Antibody binding specifically to N-terminal region of lysyl-tRNA synthetase exposed on cell membrane

- Zymedi Co., Ltd.

The present invention relates to an antibody binding specifically to an N-terminal region of lysyl-tRNA synthetase which is exposed on the cell membrane and a use thereof.

Skip to: Description  ·  Claims  ·  References Cited  · Patent History  ·  Patent History
Description
RELATED APPLICATION DATA

This application is a National Stage Application under 35 U.S.C. 371 of co-pending PCT application number PCT/KR2018/003594 designating the United States and filed Mar. 27, 2018; which claims the benefit of Korean Patent Application No. 10-2017-0038775 filed Mar. 27, 2017, Korean Patent Application No. 10-2017-0118890 filed Sep. 15, 2017 and Korean Patent Application No. 10-2017-0118917 filed Sep. 15, 2017, the entire specifications of which are incorporated herein by reference in their entireties.

SEQUENCE LISTING

The instant application contains a Sequence Listing which has been filed electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Dec. 12, 2019, is named 009041_00002_US_SL.txt and is 232,975 bytes in size.

TECHNICAL FIELD

The present invention relates to an antibody or fragment thereof specifically binding to an extracellularly exposed lysyl-tRNA synthetase N-terminal region and use thereof and, more specifically, to an antibody or fragment thereof having particular complementary determining region (CDR) sequences defined in the present specification and specifically binding to an epitope containing the sequence of SEQ ID NO: 97 in the lysyl-tRNA synthetase (KRS) N-terminus, to use of the antibody or fragment thereof for inhibition of cancer metastasis, to use of the antibody or fragment thereof for cancer diagnosis, and to a pharmaceutical composition for the prevention or treatment of an immune cell migration-related disease.

BACKGROUND ART

Recent studies have established that human lysyl-tRNA synthetase (KPS) generally present in the cytosol translocates to the plasma membrane (cell membrane) to interact with a 67-kDa laminin receptor (67LR) present on the plasma membrane, thereby promoting the migration of tumor (or cancer) cells to affect cancer metastasis (Dae Gyu Kim et al., Chemical inhibition of prometastatic lysyl-tRNA synthetaselaminin receptor interaction, Nat Chem Biol. 2014 January; 10(1): 2934, Dae Gye Kim et. al. Interaction of two translational components, lysyl-tRNA synthetase and p40/37LRP, in plasma membrane promotes laminin-dependent cell migration, FASEB J. (2012)26, 4142-4159). Human KRS (Genbank Accession No. NP_005539.1, etc) comprises an N-terminal extension (1-72), an anticodon-binding domain (73-209), and a catalytic domain (220-597). Human KRS is an enzyme essential for protein synthesis, and normally resides within the multi-tRNA synthetase complex (MSC) in the cytosol. However, after the introduction of laminin signal, p38 MAPK phosphorylates KRS at the T52 residues, and KRS translocates to the cell membrane, where KRS protects 67LR from ubiquitin-mediated degradation. It has also been reported that KRS translocated to the cell membrane accelerates cancer metastasis by stabilizing and interacting with 67LR associated with cancer metastasis.

Meanwhile, immune cells are involved in a primary defense mechanism in the body, but excessive activation of immune cells has been recently reported as one of main pathogeneses. Increased mobility of immune cells are normally observed upon the activation of inflammatory immune cells, and specifically, such immune cell migration and invasion are reported to be closely involved in disease pathology in the following diseases.

For instance, a cardiovascular disease whose lesions occur in the heart and major arteries, includes atherosclerosis and a coronary artery disease (Ross R et al., New Engl J Med, 1999:340(2):115-26, Poli G et al., Redox Biol 2013; 1(1):125-30, Libby P et al., Circulation 2002; 5; 105(9):1135-43). Atherosclerosis is an inflammatory disease triggered by cholesterol, and is caused by atheroma composed of cholesterol deposited on the inner membrane of an artery and immune cells migrating from the blood to the inside of an artery. That is, atheroma is formed by migration of immune cells, such as monocytes, to a site where oxidized cholesterol cause inflammation. The formation of atheroma roughens the interior surface of blood vessels and thickens the wall of blood vessels, and thus the inner diameter of the blood vessels becomes narrowed, resulting in circulatory disturbances. The bursting of fibrous membranes surrounding atheroma causes thrombi in the blood vessels and bleeding in atheroma, and thus the inner diameter of the blood vessels becomes rapidly narrowed or the blood vessels become blocked. This occurs mainly in blood vessels supplying blood to the heart, blood vessels supplying blood to the brain, blood vessels supplying blood to kidneys, and peripheral blood vessels, thereby causing an ischemic heart disease, an ischemic cerebrovascular disease (stroke), kidney failure, and a limb ischemic arterial disease. It has been known in the past that CC chemokine ligand 2 (CCL2, MCP-1), which causes an inflammatory response by inducing the migration of monocytes, plays an important role in the occurrence and development of such cardiovascular diseases, and therefore, new measures to treat such cardiovascular diseases by inhibiting the action of CCL2 and the resultant migration of monocytes have been suggested (Gu L et al., Mol Cell, 1998; 2(2):275-81, Aiello R J et al., Arterioscler Thromb Vasc Biol 1999; 19(6):1518-25, Gosling J1 et al., Clin Invest 1999; 103(6):773-8, Harrington J R et al., Stem Cells 2000; 18(1):65-6, Ikeda U et al., Clin Cardiol 2002; 25(4):143-7).

Pulmonary arterial hypertension (PAH) is classified as Group 1 in the clinical classification system (ESC Guidelines, European Heart Journal 2015) of the World Health Organization (WHO), and is a rare disease clinically characterized by difficulty in breathing, an increase in mean pulmonary artery pressure (mPAP, mPAP>25 mm Hg), and right ventricular dysfunction. Several pre-existing factors, such as heredity, infection, and related diseases, are involved in such pulmonary arterial hypertension, but the immune response resulting from endothelial cell injury has been known to act as a key pathological factor (Huertas et al., Circulation, 129:1332-1340, 2014). As for such a phenomenon, a series of processes according to the invasion and dysfunction of immune cells has been known to be deeply associated with pathological phenomena, and especially, the interaction between immune cells and vascular endothelial cells is known to be important in PAH. It has also been reported that the invasion of monocytes and macrophages accelerates the progress of Alport syndrome.

In fibrosis-related diseases, the continued (chronic) inflammatory responses activate the wound-healing program, leading to fibrosis. After tissue injury, inflammatory immune cells, such as monocytes/macrophages, neutrophils, eosinophils, and mast cells, invade the injured site rapidly while being activated, and secrete various cytokines, which in turn activate surrounding fibroblasts, epithelial cells, or smooth muscle cells into myoblast type cells, and these myoblast type cells produce and secrete extracellular matrix proteins in large quantities, ultimately causing the accumulation of extracellular matrix proteins in large quantities, and resulting in scar formation and tissue fibrosis or hypertrophy (Gurtner G C et al., Trends Cell Biol. 15: 599-607, 2005). This pathology is one of the fundamental causes of: scar formation in skin tissues, caused by skin wounds due to cuts, burns, bedsores, and the like; or sclerosing fibrosis of liver, kidney, vascular, and pulmonary tissues. Fibrosis is also shown to be a major pathological characteristic in chronic autoimmune diseases, such as scleroderma, rheumatoid arthritis, Crohn's disease, ulcerative colitis, myelofibrosis, and systemic lupus erythematosus. It has also been known that the activation of inflammatory immune cells contribute to pathology in atopic diseases, asthma, COPD, psoriasis, kelloid, proliferative retinopathy, and the like.

Especially in the wound-healing program, fibroblasts activated into myoblast type cells are called myofibroblasts. Since myofibroblasts are at the center of all the disease pathologies associated with fibrosis, eliminating molecular or immunological mechanisms inducing the activity of myofibroblasts is a key element of disease treatment. It has been widely known that many types of innate immunity or adaptive immunity are important in the activation and differentiation of fibroblasts, and therefore, eliminating an inflammatory response in the wound site is a key factor in stopping tissue remodeling into fibrosis and maintaining normal tissue morphology. However, since the inflammatory response is not easily eliminated in practice, understanding the mechanisms of innate and adaptive immunity to find key mediators is important in slowing fibrosis.

In some cases, monocytes, macrophages, and the like contribute to wound healing, but secrete reactive oxygen, nitrogen, and the like, and thus have harmful effects on surrounding cells. Therefore, monocytes and macrophages, if not rapidly removed, cause more tissue injury, resulting in fibrosis. Therefore, restricting monocytes and macrophages, which respond first in the early stages of the disease, is considered a therapeutic strategy for various chronic inflammation- and fibrotic-related diseases.

It has been known that when the wound healing mechanism triggers a fibrosis response, the platelet-derived growth factor (PDGF) associated with hemagglutination recruits other inflammatory immune cells into the wound site and TGF-β1 accelerates extracellular matrix synthesis from local fibroblasts. It has been however reported that the factors involved in hemagglutination induce fibrosis even when the factors are deficient.

Meanwhile, the fact that Myc-KRS41-597 (ΔN) with a deletion of 40 terminal residues in N-terminal extension (N-ext) is not localized on the plasma membrane indicates that the KRS N-ext region is an essential region in the translocation of KRS to the cell membrane. As for cancer metastasis, specifically, the KRS N-ext region has been known to be involved in the binding of KRS and 67LR in the interaction thereof. To use this fact for therapeutic or diagnostic purposes, it is necessary to specifically target a particular site (especially, KRS N-ext) in the KRS protein according to the characteristics of several domains constituting the KRS protein.

However, despite the importance of aminoacyl-tRNA synthetases (ARSs) including KRS as biomarkers, ARSs are similar in view of the protein structure, and thus the antibodies obtained via immunization of animals with a ARS protein show a cross reactivity, for example, binding with other ARSs, and in many cases, high-sensitive antibodies are not even produced.

In the diseases caused by excessive activation of immune cells as mentioned above, target factors for preventing the translocation (and invasion) of immune cells have been conventionally suggested, and attempts have been made to devise therapeutic methods to treat diseases regulating the target factors, but respective limitations thereof are being reported. Therefore, for effective disease treatment, it is still a critical challenge to establish what the key mediator is and what strategy will control the key mediator, in the mitigation of immune cells.

DETAILED DESCRIPTION OF THE INVENTION Technical Problem

While studying to construct an antibody specifically binding to an extracellularly exposed KRS N-terminal region, the present inventors verified that antibodies having particular complementary determining region (CDR) sequences defined in the present specification showed very high binding specificity and affinity to the KRS N-terminal region as well as inhibited cancer metastasis in vivo. Furthermore, the present inventors verified that an increase in KRS level in the cellular membrane of immune cells (monocytes/macrophages) is an important pathological phenomenon in immune cell migration- and invasion-related diseases, and thus KRS has a particular correlation with laminin (especially, laminin subtype α4β2γ1), and verified that KRS N-terminus binding antibodies provided in the present invention reduced the KRS level increased on the cell membrane of immune cells and actually inhibited the migration and infiltration of immune cells, and thus had an effect of treating related diseases, and therefore the present inventors completed the present invention.

Therefore, an aspect of the present invention is to provide an antibody or fragment thereof specifically binding to an epitope containing the sequence of SEQ ID NO: 97 in the lysyl-tRNA synthetase (KRS) N-terminus.

Another aspect of the present invention is to provide a polynucleotide encoding the antibody or fragment thereof of the present invention, a recombinant expression vector comprising the polynucleotide, and a cell transformed with the recombinant vector.

Still another aspect of the present invention is to provide a method for producing an antibody or fragment thereof specifically binding to an extracellularly exposed lysyl-tRNA synthetase (KRS) N-terminal region, the method comprising: (a) transforming host cells with the recombinant expression vector; (b) incubating the transformed host cells to produce an antibody or fragment thereof; and (c) collecting the antibody or fragment thereof produced in the host cells.

Still another aspect of the present invention is to provide a pharmaceutical composition comprising the antibody or fragment thereof of the present invention as an active ingredient for inhibition of cancer metastasis.

Still another aspect of the present invention is to provide a pharmaceutical composition consisting of the antibody or fragment thereof of the present invention for inhibition of cancer metastasis.

Still another aspect of the present invention is to provide a pharmaceutical composition essentially consisting of the antibody or fragment thereof of the present invention for inhibition of cancer metastasis.

Still another aspect of the present invention is to provide a composition comprising the antibody or fragment thereof of the present invention as an active ingredient for cancer diagnosis.

Still another aspect of the present invention is to provide a composition consisting of the antibody or fragment thereof of the present invention for cancer diagnosis.

Still another aspect of the present invention is to provide a composition essentially consisting of the antibody or fragment thereof of the present invention for cancer diagnosis.

Still another aspect of the present invention is to provide a pharmaceutical composition comprising the antibody or fragment thereof of the present invention as an active ingredient for the prevention or treatment of an immune cell migration-related disease.

Still another aspect of the present invention is to provide a pharmaceutical composition consisting of the antibody or fragment thereof of the present invention for the prevention or treatment of an immune cell migration-related disease.

Still another aspect of the present invention is to provide a pharmaceutical composition essentially consisting of the antibody or fragment thereof of the present invention for the prevention or treatment of an immune cell migration-related disease.

Still another aspect of the present invention is to provide use of the antibody or fragment thereof of the present invention for preparing an agent for inhibition of cancer metastasis.

Still another aspect of the present invention is to provide a method for inhibiting cancer metastasis in a subject in need thereof, the method comprising administering the antibody or fragment thereof of the present invention to the subject in an amount effective for inhibiting cancer metastasis.

Still another aspect of the present invention is to provide use of the antibody or fragment thereof of the present invention for preparing an agent for cancer diagnosis.

Still another aspect of the present invention is to provide a method for diagnosing cancer in a subject in need thereof, the method comprising administering the antibody or fragment thereof of the present invention to the subject in an amount effective for diagnosing cancer.

Still another aspect of the present invention is to provide use of the antibody or fragment thereof of the present invention for preparing an agent for the treatment of an immune cell migration-related disease.

Still another aspect of the present invention is to provide a method for treating an immune cell migration-related disease in a subject in need thereof, the method comprising administering the antibody or fragment thereof of the present invention to the subject in an amount effective for treating an immune cell migration-related disease.

Technical Solution

In accordance with an aspect of the present invention, there is provided an antibody or fragment thereof specifically binding to an epitope containing the sequence of SEQ ID NO: 97 in the lysyl-tRNA synthetase (KRS) N-terminus.

In accordance with another aspect of the present invention, there is provided a polynucleotide encoding the antibody or fragment thereof of the present invention, a recombinant expression vector comprising the polynucleotide, and a cell transformed with the recombinant vector.

In accordance with still another aspect of the present invention, there is provided a method for producing an antibody or fragment thereof specifically binding to an extracellularly exposed lysyl-tRNA synthetase (KRS) N-terminal region, the method comprising: (a) transforming host cells with the recombinant expression vector; (b) incubating the transformed host cells to produce an antibody or fragment thereof; and (c) collecting the antibody or fragment thereof produced in the host cells.

In accordance with still another aspect of the present invention, there is provided a pharmaceutical composition comprising the antibody or fragment thereof of the present invention as an active ingredient for inhibition of cancer metastasis.

In accordance with still another aspect of the present invention, there is provided a pharmaceutical composition consisting of the antibody or fragment thereof of the present invention for inhibition of cancer metastasis.

In accordance with still another aspect of the present invention, there is provided a pharmaceutical composition essentially consisting of the antibody or fragment thereof of the present invention for the inhibition of cancer metastasis.

In accordance with still another aspect of the present invention, there is provided a composition comprising the antibody or fragment thereof of the present invention as an active ingredient for cancer diagnosis.

In accordance with still another aspect of the present invention, there is provided a composition consisting of the antibody or fragment thereof of the present invention for cancer diagnosis.

In accordance with still another aspect of the present invention, there is provided a composition essentially consisting of the antibody or fragment thereof of the present invention as an active ingredient for cancer diagnosis.

In accordance with still another aspect of the present invention, there is provided a pharmaceutical composition comprising the antibody or fragment thereof of the present invention as an active ingredient for the prevention or treatment of an immune cell migration-related disease.

In accordance with still another aspect of the present invention, there is provided a pharmaceutical composition consisting of the antibody or fragment thereof of the present invention for the prevention or treatment of an immune cell migration-related disease.

In accordance with still another aspect of the present invention, there is provided a pharmaceutical composition essentially consisting of the antibody or fragment thereof of the present invention for the prevention or treatment of an immune cell migration-related disease.

In accordance with still another aspect of the present invention, there is provided use of the antibody or fragment thereof of the present invention for preparing an agent for inhibition of cancer metastasis.

In accordance with still another aspect of the present invention, there is provided a method for inhibiting cancer metastasis in a subject in need thereof, the method comprising administering the antibody or fragment thereof of the present invention to the subject in an amount effective for inhibiting cancer metastasis.

In accordance with still another aspect of the present invention, there is provided use of the antibody or fragment thereof of the present invention for preparing an agent for cancer diagnosis.

In accordance with still another aspect of the present invention, there is provided a method for diagnosing cancer in a subject in need thereof, the method comprising administering the antibody or fragment thereof of the present invention to the subject in an amount effective for diagnosing cancer.

In accordance with still another aspect of the present invention, there is provided use of the antibody or fragment thereof of the present invention for preparing an agent for treatment of an immune cell migration-related disease.

In accordance with still another aspect of the present invention, there is provided a method for treating an immune cell migration-related disease in a subject in need thereof, the method comprising administering the antibody or fragment thereof of the present invention to the subject in an amount effective for treating an immune cell migration-related disease.

Hereinafter, the present invention will be described in detail.

As used herein, the term “extracellularly exposed lysyl-tRNA synthetase (KRS) N-terminal region” refers to a particular sequence exposed to the extracellular space or on the surface of the cell membrane when KM produced in cells is translocated to the cell membrane (or plasma membrane), and may normally refer to a partial or full-length sequence of a 1- to 72-amino acid region in the KRS N-terminus. In addition, there is sequence similarity across species in the KRS N-terminal region, and especially, the KRS N-terminal region may contain the amino acid sequence defined by SEQ ID NO: 97. Preferably, the KRS N-terminal region contains the sequence defined by SEQ ID NO: 75 for humans, the sequence defined by SEQ ID NO: 113 for mice, and the sequence defined by SEQ ID NO: 114 for rats.

As used herein, the term “KRS” refers to the full-length polypeptide known as lysyl-tRNA synthetase or any KRS fragment sequence comprising the N-terminal region. As described above, the antibodies or fragments thereof according to the present invention specifically detect the extracellularly exposed KRS N-terminal region, and thus also can detect the foregoing KRS full-length polypeptide or any KRS fragment sequence containing the N-terminal region. The specific sequence of KRS is not particularly limited as long as the sequence contains the polypeptide defined by SEQ ID NO: 75 and is known as lysyl-tRNA synthetase in the art. For instance, KRS of the present invention includes: a sequence derived from a human (Homo sapiens) and known as NCBI (Genbank) Accession No. NP_005539.1 or the like; a sequence derived from a mouse (Mus musculus) and known as NCBI (Genbank) Accession No. NP_444322.1 or the like; and a sequence derived from a rat (Rattus norvegicus) and known as NCBI (Genbank) Accession No. XP 006255692.1 or the like, and besides, reference may be made to the following sequence information, but is not limited thereto: XP_005004655.1 (guinea-pig: Cavia porcellus), XP_021503253.1 (gerbil, Meriones unguiculatus), XP_002711778.1 (rabbit, Oryctolagus cuniculus), XP_536777.2 (dog, Canis lupus familiaris), XP_003126904.2 (swine, Sus scrofa), XP_011755768.1 (monkey, Macaca nemestrina), XP_008984479.1 (marmoset, Callithrix jacchus), XP_019834275.1 (cow, Bos indicus), and XP_511115.2 (chimpanzee, Pan troglodytes). Most preferably, KRS may be a polypeptide consisting of the amino acid sequence defined by SEQ ID NO: 76 (Genbank Accession No. NP_005539.1).

In the present invention, the antibody is also called immunoglobulin (Ig) and is a generic term for proteins that are involved in biological immunity by selectively acting on antigens. A whole antibody found in nature usually consists of two pairs of light chain (LC) and heavy chain (HC), each of which is a polypeptide composed of several domains, or has two pairs of HC/LC as a basic unit. There are five types of heavy chains constituting mammalian antibodies, which are denoted by the Greek letters: α, δ, ε, γ, and μ, and different types of heavy chains constitute different types of antibodies: IgA, IgD, IgE, IgG and IgM, respectively. There are two types of light chains constituting mammalian antibodies, which are denoted by λ and κ.

The heavy and light chains of antibodies are structurally divided into a variable region and a constant region according to the variability of amino acid sequence. The constant region of the heavy chain is composed of three or four heavy chain constant regions, such as CH1, CH2, and CH3 (IgA, IgD, and IgG antibodies) and CH4 (IgE and IgM antibodies), according to the type of antibody, and the light chain has one constant region CL. The variable regions of the heavy and light chains are each composed of one domain of a heavy chain variable region (VH) or a light chain variable region (VL). The light chain and the heavy chain are linked to each other by one covalent disulfide linkage while variable regions and constant regions thereof are arranged in parallel, and two heavy chain molecules, which are linked with the light chains, are linked to each other by two covalent disulfide linkages, thereby forming a whole antibody. The whole antibody specifically binds to an antigen through the variable regions of the heavy and light chains. The whole antibody is composed of two pairs of heavy and light chains (HC-LC), and thus one whole antibody molecule has divalent mono-specificity in which one whole antibody molecule binds to two same antigens through two variable regions.

The variable regions of the antibody, which comprise antigen-binding sites, are each divided into framework regions (FRs) with low sequence variability and complementary determining regions (CDRs), which are hypervariable regions with high sequence variability. In VH and VL, three CDRs and four FRs are arranged in the order of FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4 in a direction from the N-terminal to the C-terminal. CDRs, which have the highest sequence variability in the variable regions of the antibody, are sites that directly bind to an antigen, and are very important in antigen specificity of the antibody.

The present invention provides an antibody or fragment thereof specifically binding to an epitope containing the sequence of SEQ ID NO: 97 in the lysyl-tRNA synthetase (KRS) N-terminus.

As used herein, the “epitope” refers to a protein determinant capable of specifically binding to an antibody. An epitope is usually composed of surface groups of molecules, such as amino acids or sugar side chains, and usually have specific three-dimensional structural characteristics as well as specific charge characteristics. Conformational and non-conformational epitopes are distinguished from each other in that the binding to the conformational epitopes but not the non-conformational epitopes is lost in the presence of denaturing solvents. An epitope may comprise amino acid residues directly involved in the binding (also called immunogenic component of the epitope) and other amino acid residues not directly involved in the binding, for example, amino acid residues effectively blocked by the specific antigen binding peptide (in other words, the amino acid residue being within the footprint of the specific antigen binding peptide).

Preferably, the epitope is a site to which the N3 monoclonal antibody of the present invention derived from the KRS N-terminal sequence binds, and the specific sequence thereof is not particularly limited as long as the sequence is a consecutive region comprising amino acids (klsknelkrrlka) defined by SEQ ID NO: 97, and may usually consist of a 13-52 amino acid sequence, more preferably, a 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, or 42 amino acid sequence, comprising the amino acid sequence of SEQ ID NO: 97.

Preferably, the epitope of the present invention may include the amino acid sequences defined by SEQ ID NO: 75, SEQ ID NO: 98, SEQ ID NO: 99, SEQ ID NO: 100, and SEQ ID NO: 101, which are derived from the human KRS N-terminus; the amino acid sequences defined by SEQ ID NO: 113, SEQ ID NO: 103, SEQ ID NO: 104, SEQ ID NO: 105, and SEQ ID NO: 106, which are derived from the mouse KRS N-terminus; and the amino acid sequences defined by SEQ ID NO: 114, SEQ ID NO: 108, SEQ ID NO: 109, SEQ ID NO: 110, and SEQ ID NO: 111, which are derived from the rat KRS N-terminus. The epitope may be more preferably the amino acid sequence at positions 15 to 29 in the human KRS N-terminal region defined by SEQ ID NO: 75 (SEQ ID NO: 75, SEQ ID NO: 98, SEQ ID NO: 99, SEQ ID NO: 100, and SEQ ID NO: 101), and most preferably the amino acid sequence at positions 15 to 42 in the human KRS N-terminal region defined by SEQ ID NO: 75 (SEQ ID NO: 101).

The “antibody or fragment thereof specifically binding to an extracellularly exposed KRS N-terminal region” provided in the present invention comprises:

a heavy chain variable region (VH) comprising: heavy chain complementary determining region 1 (CDR1) containing the amino acid sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 13, SEQ ID NO: 25, and SEQ ID NO: 37; heavy chain complementary determining region 2 (CDR2) containing the amino acid sequence selected from SEQ ID NO: 3, SEQ ID NO: 15, SEQ ID NO: 27, and SEQ ID NO: 39; and heavy chain complementary determining region 3 (CDR3) containing the amino acid sequence selected from SEQ ID NO: 5, SEQ ID NO: 17, SEQ ID NO: 29, and SEQ ID NO: 41; and

a light chain variable region (VL) comprising: light chain complementary determining region 1 (CDR1) containing the amino acid sequence selected from the group consisting of SEQ ID NO: 7, SEQ ID NO: 19, SEQ ID NO: 31, and SEQ ID NO: 43; light chain complementary determining region 2 (CDR2) containing the amino acid sequence selected from SEQ ID NO: 9, SEQ ID NO: 21, SEQ ID NO: 33, and SEQ ID NO: 45; and light chain complementary determining region 3 (CDR3) containing the amino acid sequence selected from SEQ ID NO: 11, SEQ ID NO: 23, SEQ ID NO: 35, and SEQ ID NO: 47.

The antibodies composed of the CDR sequences have excellent ability to specifically bind to the extracellularly exposed KRS N-terminal region. This feature is well described in the examples of the present specification. In an example of the present invention, to construct scFv fragments specifically binding to the extracellularly exposed KRS N-terminal region, a total of five experimental steps starting from primary screening through scFv phage library screening to indirect ELISA (secondary screening), western blotting (tertiary screening), immunoprecipitation (quaternary screening), and immunofluorescent staining (quinary screening) were performed to select scFv fragments showing high binding specificity and binding affinity in view of KRS N-terminal binding. A total of 1920 scFv clones were selected in the primary screening through scFv phage library screening, but four types of fragments, N3 scFv, N5 scFv, N7 scFv, and N9 scFv, which have the highest specificity, were finally selected through the five steps of screening. In addition, the scFv fragments were converted into IgG antibodies, thereby constructing N3 IgG, N5 IgG, N7 IgG, and N9 IgG antibodies, and these antibodies were also verified to show high binding specificity in view of KRS N-terminal binding.

The antibodies or fragments thereof specifically binding to the extracellularly exposed KRS N-terminal region according to the present invention are antibodies having the following CDR conformations of heavy and light variable regions, wherein (i), (ii), (iii), and (iv) below indicate CDR combinations of N3, N5, N7, and N9 antibodies in respective examples:

(1) a heavy chain variable region comprising heavy chain complementary determining region 1 containing the amino acid sequence defined by SEQ ID NO: 1, heavy chain complementary determining region 2 containing the amino acid sequence defined by SEQ ID NO: 3, and heavy chain complementary determining region 3 containing the amino acid sequence defined by SEQ ID NO: 5, and a light chain variable region comprising light chain complementary determining region 1 containing the amino acid sequence defined by SEQ ID NO: 7, light chain complementary determining region 2 containing the amino acid sequence defined by SEQ ID NO: 9, and light chain complementary determining region 3 containing the amino acid sequence defined by SEQ ID NO: 11;

(2) a heavy chain variable region comprising heavy chain complementary determining region 1 containing the amino acid sequence defined by SEQ ID NO: 13, heavy chain complementary determining region 2 containing the amino acid sequence defined by SEQ ID NO: 15, and heavy chain complementary determining region 3 containing the amino acid sequence defined by SEQ ID NO: 17, and a light chain variable region comprising light chain complementary determining region 1 containing the amino acid sequence defined by SEQ ID NO: 19, light chain complementary determining region 2 containing the amino acid sequence defined by SEQ ID NO: 21, and light chain complementary determining region 3 containing the amino acid sequence defined by SEQ ID NO: 23;

(3) a heavy chain variable region comprising heavy chain complementary determining region 1 containing the amino acid sequence defined by SEQ ID NO: 25, heavy chain complementary determining region 2 containing the amino acid sequence defined by SEQ ID NO: 27, and heavy chain complementary determining region 3 containing the amino acid sequence defined by SEQ ID NO: 29, and a light chain variable region comprising light chain complementary determining region 1 containing the amino acid sequence defined by SEQ ID NO: 31, light chain complementary determining region 2 containing the amino acid sequence defined by SEQ ID NO: 33, and light chain complementary determining region 3 containing the amino acid sequence defined by SEQ ID NO: 35; and

(4) a heavy chain variable region comprising heavy chain complementary determining region 1 containing the amino acid sequence defined by SEQ ID NO: 37, heavy chain complementary determining region 2 containing the amino acid sequence defined by SEQ ID NO: 39, and heavy chain complementary determining region 3 containing the amino acid sequence defined by SEQ ID NO: 41, and a light chain variable region comprising light chain complementary determining region 1 containing the amino acid sequence defined by SEQ ID NO: 43, light chain complementary determining region 2 containing the amino acid sequence defined by SEQ ID NO: 45, and light chain complementary determining region 3 containing the amino acid sequence defined by SEQ ID NO: 47.

Most preferably, the antibodies or fragments thereof according to the present invention are characterized by comprising the following heavy chain and light chain variable regions: In the antibodies or fragments thereof, the heavy chain variable region contains the amino acid sequence selected from the group consisting of SEQ ID NO: 49 (N3 SEQ ID NO: 53 (N5 SEQ ID NO: 57 (N7 VH), and SEQ ID NO: 61 (N9 VH), and the light chain variable region contains the amino acid sequence selected from the group consisting of SEQ ID NO: 51 (N3 VL), SEQ ID NO: 55 (N5 VL), SEQ ID NO: 59 (N7 VL), and SEQ ID NO: 63 (N9 VL).

The antibody comprising the heavy chain variable region (VH) and the light chain variable region (VL) may be an antibody comprising a heavy chain containing the amino acid sequence selected from the group consisting of SEQ ID NO: 77, SEQ ID NO: 81, SEQ ID NO: 85, and SEQ ID NO: 89 and a light chain containing the amino acid sequence selected from the group consisting of SEQ ID NO: 79, SEQ ID NO: 83, SEQ ID NO: 87, and SEQ ID NO: 91.

Most preferably, the antibodies may be antibodies comprising: a heavy chain containing the amino acid sequence defined by SEQ ID NO: 77 and a light chain containing the amino acid sequence defined by SEQ ID NO: 79; a heavy chain containing the amino acid sequence defined by SEQ ID NO: 81 and a light chain containing the amino acid sequence defined by SEQ ID NO: 83; a heavy chain containing the amino acid sequence defined by SEQ ID NO: 85 and a light chain containing the amino acid sequence defined by SEQ ID NO: 87; and a heavy chain containing the amino acid sequence defined by SEQ ID NO: 89 and a light chain containing the amino acid sequence defined by SEQ ID NO: 91.

The “antibody specifically binding to the extracellularly exposed KRS N-terminal region” according to the present invention is not limited to the type thereof as long as the antibody has the above CDR combinations or VH and VL combinations. As a specific example, the antibody may be selected from the group consisting of IgG, IgA, IgM, IgE, and IgD antibodies, and may be preferably an IgG antibody.

The antibodies of the present invention may be monoclonal antibodies or polyclonal antibodies as long as the antibodies have the above CDR combinations or VH and Vt combinations that specifically bind to the KRS N-terminal region, but are preferably monoclonal antibodies, which are a group of antibodies each having substantially identical amino acid sequences in heavy and light chains.

The antibody of the present invention may be derived from any animals including mammals including humans, and birds, and may be preferably derived from humans. However, the antibody of the present invention may be a chimeric antibody including a portion of the antibody derived from humans and a portion of the antibody derived from a different species of animal. That is, the present invention includes all of chimeric antibodies, humanized antibodies, and human antibodies, and may be preferably human antibodies.

In addition, the fragment of the antibody of the present invention refers to an antibody fragment that retains antigen-specific binding ability of a whole antibody. Preferably, the fragment retains at least 20%, 50%, 70%, 80%, 90%, 95%, or 100% of the KRS N-terminal binding affinity of the mother antibody. Specifically, the fragment may be in the form of Fab, F(ab)2, Fab′, F(ab′)2, Fv, diabody, scFv, or the like.

Fab (fragment, antigen-binding) is an antigen-binding fragment of an antibody, and is composed of a heavy chain and a light chain each consisting of one variable domain and one constant domain. F(ab′)2 is a fragment produced by pepsin hydrolysis of an antibody, and F(ab′)2 has a form in which two Fab molecules are linked via disulfide bonds at the heavy-chain hinge region. F(ab′) is a monomeric antibody fragment in which a heavy-chain hinge is added to a Fab separated from F(ab′)2 fragment by the reduction of disulfide bonds thereof. Fv (variable fragment) is an antibody fragment composed of only respective variable regions of the heavy and light chains. scFv (single chain variable fragment) is a recombinant antibody fragment in which a heavy chain variable region (VH) and a light chain variable region (VL) are linked to each other via a flexible peptide linker. The diabody refers to a fragment in which VH and VL of scFv are linked by a very short linker and thus cannot be bound to each other, and bind to VL and VH of another scFv in the same form, respectively, to form a dimer.

For the purposes of the present invention, the fragment of the antibody is not limited to the structure or conformation thereof as long as the fragment of the antibody retains binding specificity to the KRS N-terminal region, but may be preferably scFv. The scFv according to the present invention has a CDR conformation or VH and VL, conformation specific to the KRS N-terminal region, and the sequence thereof is not particularly limited as long as the C-terminal of VH and the N-terminal of VL are linked through a linker. The linker is not particularly limited to the type thereof as long as it is known as a linker applied to scFv in the art, but may be a peptide containing the amino acid sequence defined by SEQ ID NO: 65. Specifically, the scFv of the present invention may contain the amino acid sequence selected from the group consisting of SEQ ID NO: 67 (N3 scFv), SEQ ID NO: 69 (N5 scFv), SEQ ID NO: 71 (N7 scFv), and SEQ ID NO: 73 (N9 scFv).

The antibody or fragment thereof of the present invention may comprise a conservative amino acid substitution (also called a conservative variant of the antibody) that does not substantially change biological activity thereof.

In addition, the foregoing antibody or fragment thereof of the present invention may be conjugated to an enzyme, a fluorescent material, a radioactive material, and a protein, but is not limited thereto. Also, methods of conjugating the above materials to the antibody have been well known in the art.

The present invention provides a polynucleotide encoding the foregoing antibody or fragment thereof according to the present invention.

In the present specification, the polynucleotide may be described as an oligonucleotide or a nucleic acid, and includes: DNA or RNA analogues (e.g., peptide nucleic acids and non-naturally occurring nucleotide analogues) generated using DNA molecules (e.g., cDNA or genomic DNA), RNA molecules (e.g., mRNA), or nucleotide analogues; and hybrids thereof. The polynucleotide may be single-stranded or double-stranded.

The polynucleotide refers to a nucleotide sequence encoding an antibody composed of heavy and light chains each having a CDR conformation or VH and VL conformation specific to the KRS N-terminal region. The polynucleotide of the present invention is not particularly limited to the sequence thereof as long as the sequence encodes the antibody or fragment thereof of the present invention. The polynucleotides encoding the foregoing CDR sequences in the above-described antibodies according to the present invention are not particularly limited to the sequences thereof, but may preferably contain the nucleotide sequence defined by SEQ ID NO: 2 (heavy chain CDR1), SEQ ID NO: 4 (heavy chain CDR2), SEQ ID NO: 6 (heavy chain CDR3), SEQ ID NO: 8 (light chain CDR1), SEQ ID NO: 10 (light chain CDR2), SEQ ID NO: 12 (light chain CDR3), SEQ ID NO: 14 (heavy chain CDR1), SEQ ID NO: 16 (heavy chain CDR2), SEQ ID NO: 18 (heavy chain CDR3), SEQ ID NO: 20 (light chain CDR1), SEQ ID NO: 22 (light chain CDR2), SEQ ID NO: 24 (light chain CDR3), SEQ ID NO: 26 (heavy chain CDR1), SEQ ID NO: 28 (heavy chain CDR2), SEQ ID NO: 30 (heavy chain CDR3), SEQ ID NO: 32 (light chain CDR1), SEQ ID NO: 34 (light chain CDR2), SEQ ID NO: 36 (light chain CDR3), SEQ ID NO: 38 (heavy chain CDR1), SEQ ID NO: 40 (heavy chain CDR2), SEQ ID NO: 42 (heavy chain CDR3), SEQ ID NO: 44 (light chain CDR1), SEQ ID NO: 46 (light chain CDR2), or SEQ ID NO: 48 (light chain CDR3).

In addition, the polynucleotides encoding the foregoing VH and VL in the antibody according to the present invention are not particularly limited to the sequences thereof, but may preferably contain the nucleotide sequence defined by SEQ ID NO: 50 (VH), SEQ ID NO: 52 (VL), SEQ ID NO: 54 (VH), SEQ ID NO: 56 (VL), SEQ ID NO: 58 (VH), SEQ ID NO: 60 (VL), SEQ ID NO: 62 (VH), or SEQ ID NO: 64 (VL).

In addition, the polynucleotide encoding the fragment of the antibody may preferably contain the nucleotide sequence of any one selected from the group consisting of SEQ ID NO: 68, SEQ ID NO: 70, SEQ ID NO: 72, and SEQ ID NO: 74, which encode scFv fragments according to the present invention.

The polynucleotides encoding the antibody or fragment thereof of the present invention may be obtained by a method known in the art. For example, on the basis of DNA sequences encoding a part or the entirety of the heavy and light chains of the antibody or corresponding amino acid sequences, the polynucleotides may be synthesized by the oligonucleotide synthesis methods that are known in the art, e.g., a polymerase chain reaction (PCR) method.

The present invention provides a recombinant expression vector comprising the polynucleotide encoding the antibody or fragment thereof according to the present invention.

As used herein, the “recombinant”, used interchangeably with “genetic manipulation”, and refers to the construction of a gene in the form that does not exist in nature, by using molecular cloning experiment techniques, such as gene transformation, cleavage, or linkage.

As used herein, the term “expression” refers to the production of proteins or nucleic acids in cells.

As used herein, the term “recombinant expression vector” is a vector that can express a target protein or nucleic acid (RNA) in a suitable host cell, and refers to a gene construct comprising essential control elements that are operably linked to be capable of expressing a polynucleotide (gene) insert. The term “operably linked” refers to the functional linkage of a nucleic acid expression control sequence and a nucleic acid sequence encoding a target protein or RNA so as to perform general functions, which means the linkage therebetween so as to allow a gene to be expressed by the expression control sequence. The expression control sequence refers to a DNA sequence that controls the expression of an operably linked polynucleotide sequence in a particular host cell. Such an expression control sequence includes a promoter for transcription, any operator sequence for controlling transcription, a sequence for encoding a proper mRNA ribosomal binding site, a sequence for controlling the termination of transcription and translation, an initiation codon, a termination codon, a polyadenylation A signal, an enhancer, and the like.

The recombinant expression vector of the present invention is not particularly limited to the type thereof as long as the vector is ordinarily used in a field of cloning, and examples of the recombinant expression vector include a plasmid vector, a cosmid vector, a bacteriophage vector, and a viral vector, but are not limited thereto. Examples of the plasmid may include Escherichia coli-derived plasmids (pBR322, pBR325, pUC118, pUC119, and pET-22b(+)), Bacillus subtilis-derived plasmids (pUB110 and pTP5), and yeast-derived plasmids (YEp13, YEp24, and YCp50), and examples of the virus may include: animal viruses, such as retrovirus, adenovirus, or vaccinia virus; and insect viruses, such as baculovirus.

The recombinant expression vector according to the present invention means a gene construct that is operably linked so as to be capable of expressing, in a suitable host cell, a polynucleotide encoding the antibody or fragment thereof composed of heavy and light chains having the foregoing CDR or VH and VL conformations capable of specifically binding the KRS N-terminal region.

The polynucleotides encoding heavy and light chains of the antibody according to the present invention may be contained in separate recombinant expression vectors, respectively, or may be contained in one recombinant expression vector.

The Present Invention Provides Cells Transformed with the Above-Described Recombinant Expression Vector.

The cells of the present invention are not particularly limited to the type thereof as long as the cells can be used to express a polynucleotide encoding an antibody or a fragment thereof contained in the recombinant expression vector of the present invention. The cells (host cells) transformed with the recombinant expression vector according to the present invention may be prokaryotic cells (e.g., E. coli), eukaryotic cells (e.g., yeast or other fungi), plant cells (e.g., tobacco or tomato plant cells), animal cells (e.g., human cells, monkey cells, hamster cells, rat cells, mouse cells, or insect cells), or hybridomas derived therefrom. Preferably, the cells may be derived from mammals including humans.

Exemplary prokaryotes suitable for the present purpose include Gram-negative or Gram-positive organisms, for example, Enterobacteriaceae such as Escherichia, e.g., E. coli, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, e.g., Salmonella typhimurium, Serratia, e.g., Serratia marcescens, and Shigella, as well as Bacilli, e.g., B. subtilis and B. licheniformis, Pseudomonas, e.g., P. aeruginosa, and Streptomyces. The cells of the present invention are not particularly limited as long as the cells can express the vector of the present invention, but may be preferably E. coli.

Saccharomyces cerevisiae is most frequently used as a eukaryote for the cells of the present invention. However, a number of other genera, species, and strains can be used, but are not limited to, for example, Schizosaccharomyces pompe; Kluyveromyces hosts, such as, K lactis, K. fragilis (ATCC 12,424), K. bulgaricus (ATCC 16,045), K. wickeramii (ATCC 24,178), K. waltii (ATCC 56,500), K. drosophilarum (ATCC 36,906), K. thermotolerans, and K. marxianus; Yarrowia (EP 402,226); Pichia pastoris (EP 183,070); Candida; Trichoderma reesia (EP 244,234); Neurospora crassa; Schwanniomyces, such as Schwanniomyces occidentalis; and filamentous fungi, for example, Neurospora, Penicillium, Tolypocladium, and Aspergillus hosts, such as A. nidulans and A. niger.

The term “transformation” refers to a modification of the genotype of a host cell due to the introduction of exotic polynucleotides, and refers to an introduction of an exotic polynucleotide into a host cell regardless of a method used for the transformation. The exotic polynucleotide introduced into the host cell is incorporated into and maintained in the genome of the host cell, or is maintained without the incorporation thereinto, and the present invention includes both.

The recombinant expression vector capable of expressing the antibody or fragment thereof specifically binding to the KRS N-terminal region according to the present invention can be introduced into cells for producing the antibody or fragment thereof, by a method known in the art, for example, but is not limited to, transient transfection, microinjection, transduction, cell fusion, calcium phosphate precipitation, liposome-mediated transfection, DEAE dextran-mediated transfection, polybrene-mediated transfection, electroporation, gene gun, and known methods for introducing nucleic acids into cells, and then can transform the cells.

The present invention provides a method for preparing an antibody or fragment thereof specifically binding to an extracellularly exposed lysyl-tRNA synthetase (KRS) N-terminal region, the method comprising:

(a) transforming host cells with the recombinant expression vector;

(b) incubating the transformed host cells to produce an antibody or fragment thereof; and

(c) collecting the antibody or fragment thereof produced in the host cells.

In step (a), in order to produce the antibody or fragment thereof according to the present invention, host cells are transformed with the recombinant expression vector, in which the polynucleotide encoding the antibody or fragment thereof is operably linked.

A person skilled in the art can perform the present step by selecting a suitable transformation method according to the selected host cells and recombinant expression vector as described above. The recombinant expression vectors comprising nucleotide sequences of heavy and light chains may be co-transformed in the same host cell to allow the heavy and light chains to be expressed in one cell, or the recombinant expression vectors comprising nucleotide sequences of heavy and light chains may be transformed in separate host cells to allow the heavy and light chains to be separately expressed.

In step (b), the transformed host cells are incubated to produce polypeptides of heavy and light chains of the antibody or fragment of the antibody according to the present invention from the recombinant expression vector introduced into the host cells.

The medium composition, incubation conditions, and incubation time for incubating the host cells may be appropriately selected according to a method ordinarily used in the art. The antibody molecules produced in the host cell may be accumulated in the cellular cytoplasm, may be secreted outside the cell or in the culture medium by a suitable signal sequence, or may be targeted using a periplasm or the like. It is also preferable that the antibody according to the present invention has a functional conformation through protein refolding using a method known in the art so as to maintain binding specificity to the KRS N-terminal. As for the production of IgG type antibody, heavy and light chains may be expressed in separate cells and then contacted with each other in a separate step to constitute the whole antibody, or heavy and light chains may be expressed in the same cell to form the whole antibody inside the cell.

In step (c), the antibody or fragment thereof produced in the host cells is obtained.

A person skilled in the art can properly select and control the collection method considering characteristics of polypeptides of the antibody or fragment thereof produced in the host cells, characteristics of the host cells, the mode of expression, or the targeting or not of the polypeptide. For example, the antibody or fragment thereof secreted into the culture medium can be collected by obtaining the culture medium, in which the host cells are cultured, removing impurities through centrifugation, and the like. In order to, as necessary, excrete the antibody present in specific organelles or cytoplasm in the cells to the outside of the cells and collect the antibody, the cells may be lysed within an extent that does not affect the functional structure of the antibody or the fragment thereof. The obtained antibody may be further subjected to a process of further removing impurities and carrying out concentration, through chromatography, filtration using a filter, dialysis, or the like.

The polypeptide in the manufacturing (production) method of the present invention may be the antibody or fragment thereof itself of the present invention, and a polypeptide to which another amino acid sequence other than the antibody or fragment thereof of the present invention is further bound. In this case, the amino acid sequence may be removed from the antibody or fragment thereof of the present invention by using a method well known to a person skilled in the art.

The antibody or fragment thereof of the present invention specifically binds to the KRS N-terminal region, and thus is useful in the diagnostic analysis for detecting and quantifying KRS proteins in, for example, particular cells, tissues, or serum. Especially, the extracellularly exposed KRS N-terminal region can be specifically detected without cell lysis. Therefore, the present invention provides a method for specific detection of an extracellularly exposed lysyl-tRNA synthetase (KRS) N-terminal region, the method comprising: contacting the antibody or fragment thereof with a sample; and detecting the antibody or fragment thereof.

The detection method of the present invention may comprise a step of preparing a sample, which is to be measured for the presence or absence of KRS (or extracellularly exposed KRS N-terminal peptide) and the concentration thereof by using the antibody or fragment thereof according to the present invention (step (1)), before contacting the antibody or fragment thereof according to the present invention with the sample.

A person skilled in the art may suitably select a known protein detection method using an antibody and prepare a sample suitable for the selected method. In addition, the sample may be cells or tissues obtained by biopsy, blood, whole blood, serum, plasma, saliva, cerebrospinal fluid, or the like, which is collected from a subject to be examined for the presence or absence of cancer (especially breast cancer or lung cancer) or cancer metastasis. Examples of the protein detection method using the antibody include, but are not limited to, western blotting, immune blotting, dot blotting, immunohistochemistry, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay, competitive binding assay, immunoprecipitation, and the like. For example, for western blotting, a preparation may be made by adding a buffer suitable for electrophoresis to a sample or cell lysate, followed by boiling, and for immunohistochemistry, a treatment may be performed by immobilizing and blocking cells or tissue slices, followed by blocking.

Next, a step of contacting the antibody or fragment thereof according to the present invention with the sample prepared in the above-described step is performed (step (2)).

The antibody according to the present invention is an antibody or fragment thereof that has the above-described CDR or VH and VL conformations and specifically binds to the KRS N-terminal region, and specific types and sequence organization thereof are as described above.

The antibody or fragment thereof may be labeled with a general detectable moiety, for “detection” thereof. For instance, the antibody or fragment thereof may be labeled with a radioisotope or fluorescent label by using the technique described in literature [Current Protocols in Immunology, Volumes 1 and 2, 1991, Coligen et al., Ed. Wiley-Interscience, New York, N.Y., Pubs]. In addition, various enzyme-substrate labels are usable, and examples of the enzymatic label include: luciferase, such as Drosophila luciferase and bacterial luciferase (U.S. Pat. No. 4,737,456), luciferin, 2,3-dihydrophthalazine dionise, malate dehydrogenase, urase, peroxidase such as horseradish peroxidase (HRPO), alkaline phosphatase, β-galactosidase, glucoamylase, lysozyme, saccharide oxidase (e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase), heterocyclic oxidase (e.g., uricase and xanthine oxidase), lactoperoxidase, microperoxidase, and the like. Techniques for conjugating enzymes to antibodies are described in, for example, literature [O'Sullivan et al., 1981, Methods for the Preparation of Enzyme-Antibody Conjugates for use in Enzyme Immunoassay, in Methods in Enzym. (J. Langone & H. Van Vunakis, eds.), Academic press, N. Y., 73: 147-166]. The labels may be directly or indirectly conjugated to antibodies using various known techniques. For instance, the antibody may be conjugated to biotin, and any labels pertaining to three classes of widespread categories cited above may be conjugated to avidin or vice versa. Biotin may selectively bind to avidin, and therefore, this label may be conjugated to an antibody in such an indirect manner. Alternatively, in order to attain the indirect conjugation of a label to an antibody, the antibody may be conjugated to a small hapten (e.g., dioxin), and one of different types of labels recited above may be conjugated to an anti-hapten antibody (e.g., anti-dioxin antibody). Therefore, the indirect conjugation of a label to an antibody can be attained.

As used herein, the “contacting” is used in a general sense thereof, and refers to the mixing, binding, or touching of two or more substances. The contacting may be performed in vitro or in another container, or may be performed in situ, in vivo, in the subject, in the tissue, or in the cell.

Next, a step of detecting the antibody or fragment thereof according to the present invention from the sample after the execution of step (2) is performed (step (3)).

The “detection” is performed on a complex of the antibody or fragment thereof according to the present invention and an antigen, the complex being formed in the sample, and refers to the detection of the presence or absence of the KRS N-terminal peptide (or a protein including the peptide, for example, KRS) or the measurement (including qualitative measurement, quantitative measurement, or both) of the level of the peptide. Therefore, the detection method of the present invention may further comprise a step of removing extra antibodies or fragments thereof, which did not form the complex together with the KRS N-terminal region, after the execution of step (2) before step (3) to be described later.

When the antibody or fragment thereof used in step (2) described above contains a detectable moiety, such as fluorescence, radioactive isotope, or enzyme, which directly labels the antibody or fragment thereof, the detection may be carried out by a detection method for the corresponding moiety, known in the art. For instance, radioactivity may be measured by, for example, scintillation counting, and fluorescence may be quantified using a fluorometer.

When the antibody or fragment thereof, per se, used in step (2) described above does not contain the foregoing detectable moiety, the indirect detection using a secondary antibody labeled with fluorescence, radioactivity, enzyme, or the like may be carried out. The secondary antibody binds to the antibody or fragment thereof (primary antibody) according to the present invention.

Recent studies established that human lysyl-tRNA synthetase (KRS) present in the cytosol translocates to the plasma membrane (cell membrane) to interact with a 67-kDa laminin receptor (67LR) present on the plasma membrane, thereby promoting the migration of tumor (or cancer) cells to affect cancer metastasis (Dae Gyu Kim et al., Chemical inhibition of prometastatic lysyl-tRNA synthetaselaminin receptor interaction, Nat Chem Biol. 2014 January; 10(1): 2934.). Here, the KRS N terminal extension (N-ext) region has been known to be essential in the translocation of KRS to the cell membrane. As for cancer metastasis, specifically, the KRS N-ext region has been known to be involved in the binding of KRS and 67LR in the interaction thereof.

The antibodies and fragments thereof according to the present invention are excellent in specific binding ability to the KRS N-ext region. Actually, the antibodies and fragments thereof according to the present invention bind to the KRS N-ext region, and thus inhibit the binding (interaction) with a laminin receptor, thereby showing excellent ability to inhibit cancer metastasis. This is well described in the examples of the invention. An example in the present specification verified that as a result of administering the antibody according to the present invention into in vivo cancer metastasis models with induced cancer, the antibody of the present invention showed excellent cancer metastasis-inhibiting ability in a dose-dependent manner. Especially, the cancer metastasis inhibitory ability of the antibody of the present invention was very excellent even compared with YH16899 compound, which is known to inhibit cancer metastasis by inhibiting the interaction between the laminin receptor (67LR) and KRS.

Therefore, the present invention provides a pharmaceutical composition for inhibition of cancer metastasis and a composition for cancer diagnosis, each of the compositions comprising the foregoing antibody or fragment thereof of the present invention as an active ingredient for inhibition of cancer metastasis.

Furthermore, the present invention provides a pharmaceutical composition for inhibition of cancer metastasis and a composition for cancer diagnosis, each the compositions consisting of the foregoning antibody or fragment thereof of the present invention.

Furthermore, the present invention provides a pharmaceutical composition for inhibition of cancer metastasis and a composition for cancer diagnosis, each the compositions essentially consisting of the foregoning antibody or fragment thereof of the present invention.

The cancer is not particularly limited to the type thereof as long as the cancer is known as a malignant tumor in the art, and example thereof may be selected from the group consisting of breast cancer, large intestine cancer, lung cancer, small cell lung cancer, gastric cancer, liver cancer, blood cancer, bone cancer, pancreatic cancer, skin cancer, head or neck cancer, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, anal cancer, colon cancer, breast cancer, fallopian tube carcinoma, endometrial carcinoma, cervical cancer, vaginal cancer, vulvar carcinoma, Hodgkin's disease, esophageal cancer, small intestine cancer, endocrine cancer, thyroid cancer, parathyroid carcinoma, adrenal cancer, soft tissue sarcoma, uterine cancer, penis cancer, prostate cancer, chronic or acute leukemia, lymphocyte lymphoma, bladder cancer, kidney or ureter cancer, renal cell carcinoma, renal pelvic carcinoma, CNS tumor, primary CNS lymphoma, spinal cord tumor, brain stem glioma, and pituitary adenoma. Preferably, the cancer may be breast cancer or pulmonary cancer.

The present invention provides a pharmaceutical composition comprising the antibody or fragment thereof of the present invention as an active ingredient for the prevention or treatment of an immune cell migration-related disease.

Furthermore, the present invention provides a pharmaceutical composition consisting of the antibody or fragment thereof of the present invention for the prevention or treatment of an immune cell migration-related disease.

Furthermore, the present invention provides a pharmaceutical composition essentially consisting of the antibody or fragment thereof of the present invention for the prevention or treatment of an immune cell migration-related disease.

As used herein, the term “immune cells” preferably refers to monocytes or macrophages.

As used herein, the term “immuno cell migration-related disease” is not particularly limited to the specific type thereof as long as it is known in the art that excessive migration (and invasion) of immune cells is the main pathogenesis of the disease, and examples thereof may be selected from the group consisting of a cardiovascular disease, a fibrotic disease, a chronic inflammatory disease, and Alport syndrome.

The cardiovascular disease is not particularly limited to the following specific types of cardiovascular diseases and may be selected from the group consisting of pulmonary arterial hypertension, atherosclerosis, angina pectoris, myocardial infarction, ischemic cerebrovascular disease, arteriosclerosis, and mesenteric sclerosis.

The fibrotic disease is not particularly limited to the following specific types of the fibrotic diseases, and may be selected from the group consisting of scleroderma, rheumatoid arthritis, Crohn's disease, ulcerative colitis, myelofibrosis, pulmonary fibrosis, hepathic fibrosis, liver cirrhosis, kidney fibrosis, myofibrosis, cardiac fibrosis, systemic lupus erythematosus, hereditary fibrosis, infectious fibrosis (especially fibrosis caused by continuous infection), irritant fibrosis (fibrosis caused by repetitive exposure to irritant materials, such as tobacco and toxic materials), fibrosis caused by chronic autoimmune, fibrosis caused by antigen incompatibility during organ transplantation, fibrosis by hyperlipidemia, fibrosis by obesity, diabetic fibrosis, fibrosis by hypertension, and occlusion caused by fibrosis in stent insertion.

The chronic inflammatory disease may be selected from the group consisting of asthma, atopic dermatitis, eczema, psoriasis, osteoarthritis, gout, psoriatic arthritis, cirrhosis, nonalcoholic steatohepatitis, chronic obstructive pulmonary disease, rhinitis, diabetic retinopathy, diabetic renal failure, diabetic neuropathy, and multiple sclerosis.

The pharmaceutical composition according to the present invention may comprise the antibody or fragment thereof of the present invention alone or may further comprise at least one pharmaceutically acceptable carrier. As used herein, the term “pharmaceutically acceptable” refers to a non-toxic composition that is physiologically acceptable, does not inhibit action of an active ingredient when administered to humans, and does not normally cause an allergic response or similar responses, such as gastroenteric troubles and dizziness.

In the pharmaceutical composition according to the present invention, the antibody or fragment thereof may be administered in several oral and parental dosage forms during clinical administration. The antibody or fragment thereof, when formulated, may be prepared using a diluent or an excipient, such as a filler, an extender, a binder, a wetting agent, a disintegrant, or a surfactant, which is normally used. Solid formulations for oral administration include a tablet, a pill, a powder, granules, a capsule, a troche, and the like. These solid formulations may be prepared by mixing the antibody or fragment thereof of the present invention or a pharmaceutically acceptable salt thereof with at least one excipient, for example, starch, calcium carbonate, sucrose or lactose, or gelatin. In addition, lubricants, such as magnesium stearate and talc, may be used besides to the simple excipients. Liquid formulations for oral administration include a suspension, a solution for internal use, an emulsion, a syrup, and the like. Besides simple diluents that are frequently used, such as water and liquid paraffin, several excipients, for example, a wetting agent, a sweetener, an aroma, a preservative, and the like may be contained in the liquid formulations.

Exemplary formulations for parenteral administration include a sterile aqueous solution, a non-aqueous solvent, a suspension solvent, an emulsion, a freeze-drying agent, and a suppository. The composition for treatment of the present invention may be prepared in the form of a freeze-dried cake or an aqueous solution in order to mix and store any physiologically acceptable carrier, excipient, or stabilizer (Remington: The Science and Practice of Pharmacy, 19th Edition, Alfonso, R., ed, Mack Publishing Co. (Easton, Pa.: 1995)) and an antibody with preferable purity. The acceptable carrier, excipient, or stabilizer is non-toxic to a user at the used dose and concentration, and examples thereof include: buffers, for example, phosphoric acid, citric acid, and other organic acids; antioxidants including ascorbic acid; low-molecular weight (less than about 10 residues) polypeptides; proteins, for example, serum albumin, gelatin, or immunoglobulin; hydrophilic polymers, for example, polyvinyl pyrrolidone; amino acids, for example, glycine, glutamine, asparagine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrin; chelating agents, for example, EDT; sugar alcohols, for example, mannitol or sorbitol; salt-forming counter ions, for example, sodium; and (or) non-ionic surfactants, for example, Tween, pluronics, or polyethylene glycol (PEG).

The antibody of the present invention may be administered in a pharmaceutically effective amount to a subject fighting against cancer or an immune cell migration-related disease. As used herein, the term “pharmaceutically effective amount” refers to an amount showing a higher response compared with negative control, and preferably refers to an amount sufficient to treat cancer, an amount sufficient to inhibit cancer metastasis, and an amount sufficient to treat an immune cell migration-related disease. The total effective amount of the antibody or fragment thereof of the present invention may be administered to a patient as a single dose, or may be administered by a fractionated treatment protocol, in which multiple doses are administered for a long period of time. The dose of the antibody or fragment thereof of the present invention to the human body may be normally 0.01-100 mg/kg/week, preferably 0.1-20 mg/kg/week, and more preferably 5-10 mg/kg/week. However, as for the dose of the antibody or fragment thereof of the present invention, an effective dose thereof with respect to a patient is determined in consideration of various factors, for example, the route of administration of the pharmaceutical composition, the number of times of treatment, a patient's age, body weight, health condition, and sex, the severity of disease, the diet, and the excretion rate, and therefore, considering this fact, a person skilled in the art could determine a suitable effective amount of the antibody or fragment thereof of the present invention according to the particular use as a cancer metastasis inhibitor. The pharmaceutical composition according to the present invention is not particularly limited to the dosage form, route of administration, and administration method thereof as long as the composition shows effects of the present invention.

The route of administration of the composition of the present invention may be a known antibody administration method, for example, the injection or infusion by an intravenous, intraperitoneal, intracranial, subcutaneous, intramuscular, intraocular, intraarterial, cerebrospinal, or intralesional route, or the injection or infusion by the sustained release system described below. For example, the antibody of the present invention may be administered systemically or locally.

The pharmaceutical composition of the present invention may be used alone or in combination with surgery, hormone therapy, chemotherapy, and methods using biological response controller, for cancer prevention or treatment.

The pharmaceutical composition of the present invention may also be used alone or in combination with surgery, hormone therapy, chemotherapy, and methods using biological response controller, for prevention or treatment of an immune cell migration-related disease.

The diagnosis and prognosis of cancer (or cancer metastasis) according to the present invention may be evaluated by detecting KRS proteins (especially, extracellularly exposed KRS N-terminal region) in the biological sample, and the diagnosis and prognosis of the immune cell migration-related disease according to the present invention may be evaluated by detecting KRS proteins (especially, extracellularly exposed KRS N-terminal region) in the biological sample.

As used herein, the term “diagnosis” refers to identifying the presence or characteristics of a pathological condition. In the present invention, the diagnosis is to identify the occurrence or the likelihood (risk) of cancer or/and cancer metastasis or an immune cell migration-related disease.

The term “detection” is as described above, and the biological sample includes blood and other liquid samples having biological origins, biopsy specimens, solid tissue samples such as tissue culture, or cells derived therefrom. More specifically, examples of the biological sample may include, but are not limited to, tissues, extracts, cell lysates, whole blood, plasma, serum, saliva, ocular fluid, cerebrospinal fluid, sweat, urine, milk, ascites fluid, synovial fluid, peritoneal fluid, and the like. The sample may be obtained from animals, preferably mammals, and most preferably humans. The sample may be pre-treated before use for detection. Examples of the pretreatment may include filtration, distillation, extraction, concentration, interference ingredient deactivation, reagent addition, and the like. In addition, nucleic acids and proteins isolated from the sample may be used for detection.

The antibody or fragment thereof according to the present invention may be provided as a diagnostic kit. The kit is not particularly limited to the type thereof as long as the kit is known in the art as an assay kit that provides a peptide having an antibody or a particular binding domain as a component, and examples thereof include a kit for western blotting, ELISA, radioimmunoassay, radioimmunodiffusion, Ouchterlony immunodiffusion, rocket immunoelectrophoresis, immunohistochemistry, immunoprecipitation assay, complement fixation assay, FACS, a protein chip, or the like.

The antibody or fragment thereof of the present invention may be used in a kit, i.e., a packaged combination of reagents in predetermined amounts with instructions for performing the diagnostic assay. Where the antibody is labeled with an enzyme, the kit may include substrates and cofactors required by the enzyme (e.g., a substrate precursor which provides the chromophore or fluorophore). In addition, other additives may be included such as stabilizers, buffers (e.g., a block buffer or lysis buffer) and the like. The relative amounts of various reagents may be varied widely to provide concentrations in solution of the reagents which substantially optimize the sensitivity of the assay. The reagents may be provided as dry powders, usually lyophilized, including excipients which on dissolution will provide a reagent solution having an appropriate concentration.

The present invention provides use of the antibody or fragment thereof of the present invention for preparing an agent for the inhibition of cancer metastasis.

The present invention provides a method for inhibiting cancer metastasis in a subject in need thereof, the method comprising administering the antibody or fragment thereof of the present invention to the subject in an amount effective for inhibiting cancer metastasis.

The present invention provides use of the antibody or fragment thereof of the present invention for preparing an agent for cancer diagnosis.

The present invention provides a method for diagnosing cancer in a subject in need thereof, the method comprising administering the antibody or fragment thereof of the present invention to the subject in an amount effective for diagnosing cancer.

The present invention provides use of the antibody or fragment thereof of the present invention for preparing an agent for the treatment of an immune cell migration-related disease.

The present invention provides a method for treating an immune cell migration-related disease in a subject in need thereof, the method comprising administering the antibody or fragment thereof of the present invention to the subject in an amount effective for treating the immune cell migration-related disease.

As used herein, term “effective amount” refers to an amount to show an effect of alleviation, treatment, prevention, detection, or diagnosis of cancer or an effect of inhibiting or reducing cancer metastasis, and refers to an amount to show an effect of alleviation, treatment, prevention, detection, or diagnosis of an immune cell migration-related disease. Term “subject” may be an animal, preferably a mammal, especially an animal including a human being, and may be cells, a tissue, an organ, or the like derived from an animal. The subject may be a patient in need of the effect.

As used herein, term “treatment” broadly refers to alleviating cancer, a cancer-related disease, or an immune cell migration-related disease, and may include healing or substantially preventing such a disease or alleviating a condition of the disease, and may include alleviating, healing, or preventing one or most of the symptoms resulting from cancer or a cancer-related disease, but is not limited thereto.

As used herein, the term “comprising” is used synonymously with “containing” or “being characterized”, and does not exclude additional ingredients or steps not mentioned in the composition or method. The term “consisting of” means excluding additional elements, steps, or ingredients not otherwise specified. The term “essentially consisting of” means including the mentioned elements or steps as well as any element or step that does not substantially affect basic characteristics of the mentioned elements or steps in the scope of compositions or methods.

Advantageous Effects

The antibodies or fragments thereof according to the present invention have particular complementary determining regions (CDRs) defined in the present specification and a very excellent specific binding ability to an extracellularly exposed KRS N-terminal region. Furthermore, the antibodies or fragments thereof according to the present invention is specifically targeted to the KRS N-terminal region in vivo, and thus inhibit the interaction between the laminin receptor and the KRS N-terminal region, thereby exerting an excellent effect on the inhibition of cancer metastasis, and can control the migration of immune cells, thereby exerting a very remarkable effect on the prevention, alleviation, and treatment of an immune cell migration-related disease.

BRIEF DESCRIPTION OF THE DRAWINGS

The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.

FIG. 1 shows the results of selecting, through western blotting (WB), scFv phage clones binding to KRS full-length sequence or KRS N-terminal fragments.

FIG. 2 shows the results of selecting, through immunoprecipitation (IP), scFv phage clones binding to KRS full-length sequence or KRS N-terminal fragments.

FIGS. 3A-3B show staining of WI′ and mutant myc-KRS. FIG. 3A shows the results, wherein cells having KRS N-terminal region exposed on the cell membrane were constructed through myc-KRS T52D (active mutant) expression and it was investigated whether N3, N5, N7, and N9 scFv clones bound to the exposed region (KRS: meaning myc-KRS T52D). FIG. 3B shows that in the inactive mutant (myc-KRS T52A) or WT-KRS (laminin untreated) expression group, no detection signals were observed in spite of the treatment with N3, N5, N7, and N9 scFv clones since the KRS N-terminal region was not exposed on the cell membrane. FIG. 3C shows that when cells transformed with myc-labeled WT-KRS, T52D, and T52A were treated with laminin and scFv staining was performed, staining was observed at similar sites in the membrane localization of WT-KRS by laminin and T52D mutant and the transformed myc was also stained at the same site, but staining was not observed in T52A (scFv: green, myc: red).

FIG. 4 shows the western blotting results of confirming whether N3, N5, N7, and N9 scFv clones specifically bound to KRS N-terminus by using full-length KRS (denoted by F, SEQ ID NO: 76), a KRS fragment with deletion of amino acids at positions 1-71 in the N-terminus (denoted by 1), and a KRS fragment composed of amino acid residues 1-200 in the N-terminus (denoted by 2).

FIGS. 5A-5B show KRS binding ability. FIG. 5A shows the western blotting results of confirming the KRS binding ability of N3 IgG and N5 IgG as representatives among the antibodies of the present invention. FIG. 5B shows the immunoprecipitation results of confirming KRS binding ability of N3 IgG and N5 IgG as representatives among the antibodies of the present invention.

FIGS. 6A-6B show KRS N-terminus binding ability. FIG. 6A shows the SPR results of quantitatively confirming KRS N-terminus binding ability of N3 IgG. FIG. 6B shows the SPR results of quantitatively confirming KRS N-terminus binding ability of N5 IgG.

FIGS. 7A-7C show immunofluorescence staining. FIG. 7A shows the results, wherein the cells transformed to express WT-KRS were treated with laminin to extracellularly expose KRS N-terminal region, and then treated with the antibody of the present invention, N3 IgG, to investigate the binding of N3 IgG to the exposed region through immunofluorescence staining. FIG. 7B shows the results, wherein the cells having extracellularly exposed KRS N-terminal region were constructed through T52D-KRS (active mutant, myc tagged KRS) expression, and then treated with the antibody of the present invention, N3 IgG, to investigate the binding of N3 IgG to the exposed region through immunofluorescence staining. It was also confirmed through an experiment to impart permeability to the cell membrane that the antibody of the present invention translocates into cells and can bind with KRS protein present inside the cells. FIG. 7C shows the results, wherein the cells transformed to express WT-KRS were treated with laminin to extracellularly expose KRS N-terminal region, and then treated with the antibody of the present invention, N5 IgG, to investigate the binding of N5 IgG to the exposed region through immunofluorescence staining (N5 IgG: green).

FIG. 8 shows the MTT assay results of confirming that the antibody of the present invention N3 IgG had no cytotoxicity.

FIGS. 9A-9B show results of cell migration suppression. FIG. 9A shows the results of confirming that cell migration was suppressed by the treatment with the antibody of the present invention N3 IgG. FIG. 9B shows the results of confirming that the antibody of the present invention N3 IgG significantly suppressed cell migration in a dose-dependent manner.

FIG. 10 schematically shows an experimental schedule of construction of mouse cancer metastasis models, administration of therapeutic substance (antibody or YH16899), and observation of lung metastasis in the mouse models, in an experiment using in vivo cancer metastasis models.

FIG. 11 shows mouse lung specimens capable of confirming that caner metastasis to lungs were significantly suppressed by the administration of the antibody of the present invention, N3 IgG, in in-vivo cancer metastasis models. The degrees of progression and severity of cancer metastasis could be evaluated from the count and condition of nodules generated in the lung specimens.

FIG. 12 shows the results of confirming that the generation of lung nodules was significantly suppressed by the administration of the antibody of the present invention, N3 IgG, compared with control, in in-vivo cancer metastasis models (i.e., cancer metastasis to lungs were significantly suppressed).

FIG. 13 comparatively shows the lung tissues in control and the N3 IgG treatment group, and confirmed that a significantly large amount of laminin receptors were expressed in the metastasis nodule sites in control compared with the group treated with the antibody of the present invention.

FIG. 14 shows the lung specimens of control, YH16899 treatment group, N3 IgG treatment group, and shows the results that the generation of lung nodules was significantly suppressed in the YH16899 treatment group and the N3 IgG treatment group compared with control.

FIG. 15 shows the lung metastasis inhibition efficiency (efficiency of inhibiting lung nodule formation) of the cancer metastasis inhibitor substances according to the treatment concentration in the YH16899 treatment group and the N3 IgG treatment group.

FIG. 16 shows the SPR results of quantitatively confirming the binding ability of N3 IgG to human (h) KRS N-terminal peptide fragments (F1, F2, F3, F4, and F5) (gray bars below the sequence indicate the binding ability of N3 antibody to corresponding regions (F1 to F5), and the darker the bar, the stronger the binding ability).

FIG. 17 shows the SPR results of quantitatively confirming the binding ability of N3 IgG to KRS N-terminal peptide fragments (F1, F3, and F5) of human (h), mouse (m), and rat (r) (gray bars below the sequences indicate the binding ability of N3 antibody to corresponding regions (F1 to F5), and the darker the bar, the stronger the binding ability).

FIGS. 18A-18B show results of cell migration. FIG. 18A shows the transwell migration assay results of comparing the effects of collagen, fibronectin, and laminin on immune cell (monocyte/macrophage) migration, and provides microscopic images of migrating cells. FIG. 18B is a graph showing cell counts measured (quantified) on the microscopic images of FIG. 18A.

FIGS. 19A-19C show results of cell migration. FIG. 19A shows the transwell migration assay results of comparing the effects of various laminin subtypes (LN111, LN211, LN221, LN411, LN421, LN511, and LN521) on immune cell (monocyte/macrophage) migration, and provides microscopic images of migrating cells. FIG. 19B is a graph showing the cell count measured (quantified) on the microscopic images of FIG. 19A. FIG. 19C shows the western blotting results of confirming that KRS increased on the monocyte/macrophage membrane by LN421 treatment.

FIGS. 20A-20C show results of cell migration. FIG. 20A shows the Transwell migration assay results of comparing the inhibitory effects of the antibody of the present invention, N3 IgG, on LN421-specific monocyte/macrophage migration, and provides microscopic images of migrating cells. FIG. 20B is a graph showing the cell counts measured (quantified) on the microscopic images of FIG. 20 A. FIG. 20C shows the western blotting results of confirming that the KRS level increased by LN421 treatment in the monocyte/macrophage membrane was reduced by the treatment with the antibody of the present invention, N3 IgG.

FIGS. 21A-21B show changes in right and left ventricular end-systolic pressure. FIG. 21A shows a change in right ventricular end-systolic pressure (RVESP) by the administration of the antibody of the present invention, N3 IgG, in pulmonary arterial hypertension (PAR) models (Mock IgG: negative control, Ab 1 mpk: N3 antibody 1 mpk, Ab 10 mpk: N3 antibody 10 mpk, sildenafil: positive control). FIG. 21B shows a change in left ventricular end-systolic pressure (LVESP) by the administration of the antibody of the present invention, N3 IgG, in pulmonary arterial hypertension (PAH) models (Mock IgG: negative control, Ab 1 mpk: N3 IgG 1 mpk, Ab 10 mpk: N3 IgG 10 mpk, sildenafil: positive control).

FIG. 22 shows the IHC staining results of confirming that the administration of the antibody of the present invention, N3 IgG, reduced immune cell migration and invasion in the pulmonary arterial hypertension (PAH) models.

MODE FOR CARRYING OUT THE INVENTION

Hereinafter, the present invention will be described in detail.

However, the following examples are merely for illustrating the present invention and are not intended to limit the scope of the present invention.

Example 1

sdFv Library Screening

<1-1> Screening of scFv Phages Primary Screening

In order to select scFv antibodies specifically binding to only the KRS N-terminal region (SEQ ID NO: 75) extracellularly exposed when KRS translocates to the cell membrane by the laminin signal, in the KRS full-length sequence (SEQ ID NO: 76), phage display panning was performed using the scFv phage library derived from HA-tagged human B cells. The scFv display phage library (Library size: app. 7.6×109 Library produced by prof. Hyunbo Shim) used in the present experiment is disclosed in Korean Patent No. 10-0961392. As shown in Table 1 below, the KRS full-length sequences and KRS fragments with particular different regions of the N-terminus were used as antigen proteins for phage display panning.

To an immuno-tube containing 1 ml of 1×PBS solution, 1-10 μg of antigen proteins were added and incubated at 37° C. for 1 h at 200 rpm, thereby coating the inner surface of the tube with antigens. The antigen solution was drained, and uncoated antigens were removed by washing once with tap water. To prevent non-specific binding between antigen proteins and phages, the immuno-tube and scFv library were separately incubated with 1×PBST (0.05% tween20-containing PBS) containing 3% skim milk at room temperature for 1 h. After the skim milk was removed from the immuno-tube, scFv library was added and incubated at 150 rpm for 1 h at 37° C., thereby binding scFv phages to the antigens. After the scFv phages were incubated in the tube, unbound scFv phages were removed by washing two or five times with 1×PBST.

ScFv phages specifically binding to the respective KRS antigens were isolated within 10 min by addition of 1 ml of triethylamine (100 mM) at room temperature, and neutralized with Tris (1 M, pH 7.4). The filtered scFv phages were added to ER2537 E. coli cultured to OD<1, followed by infection with incubation at 120 rpm for 1 h and 30 min at 37° C. E. coli infected with the phages was centrifuged to partially remove the culture supernatant, followed by re-dispersion, and then spread on 15 cm-diameter agarose plate containing ampicillin and glucose (2%). The next day, 5 ml of SB medium was applied to collect all of the cells grown in the plate, and glycerol (50%) was added to 0.5 times the total volume, followed by mixing, and then the mixture was dispensed in 1 ml portions and stored at −80° C. (scFv panning stock). Then, 20 μl of the prepared stock was seeded in 20 ml of SB solution, followed by incubation, and then constructed into scFv phage library (1 ml) for the next step of phage panning by using helper phages. The above procedure for isolating phages expressing scFv specific to antigens was repeated two or three times.

<1-2> Screening of Specifically Binding scFv Antibodies Through ELISA_Secondary Screening

It was investigated through indirect ELISA whether scFv-expressed phages selected in Example <1-1> bound to the foregoing full-length KRS or N-terminal fragments.

The scFv product obtained by three rounds of panning was diluted, and applied on 10 cm-diameter agarose plate. The next day, respective colonies were selected, and incubated in 96-well plate containing 200 μl of SB medium. After the colonies were checked to grow well overall, IPTG (1 mM) was added, followed by incubation at 30° C. for 16 h, thereby inducing scFv production. The next day, the 96-well plate was centrifuged to isolate only cells, and then the cells were lysed with TES solution, followed by re-centrifugation, thereby separating only the supernatant. The obtained supernatant was subjected to indirect ELISA to select scFv specifically binding to antigens. The plates were coated with KRS full-length antigens or the N-terminal fragment antigens, respectively, incubated with the culture supernatant containing scFv, and then incubated with anti-HA-HRP antibody (Roche Applied Science) as a secondary antibody. The color development was performed using tetramethyl benzimidine (TMB, Thermo scientific), and then stopped using H2SO4 (1 M), and the absorbance was read at 450 nm using ELISA reader. ELISA was performed on control (blank) and the above-described antigens (Ag) at the same time to select only colonies with positive values. Out of the total of 1920 colonies subjected to ELISA, 93 colonies were selected as being positive (see Table 1).

TABLE 1 Antigen (KRS fragment) Full 1-20 13-36 31-46 1-29 5-34 10-38 15-42 24-49 Total Panning 288 384 384 192 192 192 96 96 96 1920 Outcomes HITS 51 8 20 2 1 1 0 8 2 93

<1-3> Sequencing

Sequences were analyzed to filter out the same Ab with overlapping CDR sequences out of 93 colonies selected through ELISA screening in Example <1-2>. Sequencing was specifically performed by the following method: After E. coli retaining scFv clones was cultured, phagemids were obtained by miniprep. The phagemids were sequenced using Omp primer (Hye young Yang, et. al., 2009, Mol. Cells 27, 225-235). The sequence thus obtained was used to verify the sequences of CDR regions of the phagemids by Bioedit program. Out of these, the clones with overlapping CDR sequences were eliminated to verify scFv clones of respective independent CDR sequences.

TABLE 2 Antigen (KRS fragment) Full 1-20 13-36 31-46 1-29 5-34 10-38 15-42 24-49 Total HITS 51 8 20 2 1 1 0 8 2 93 Different 10 8 11 1 1 1 0 4 2 38 Clones

As a result of screening antibodies with overlapping CDR sequences through sequencing, 38 scFv clones with different CDR sequences were obtained as shown in Table 2.

<1-4> Screening of Specifically Binding scFv Antibodies Through Western Blotting_Tertiary Screening

It was investigated through western blotting whether 38 scFv clones isolated in Example <1-3> specifically bound to KRS.

The scFv positive single-colony clones were incubated in 5 ml of kanamycin-containing SB medium (Bactotrytone 30 g, yeast extract 20 g, MOPS buffer 10 g/L) to start seed culture, and after incubation overnight, the culture was transferred to 500 ml of kanamycin-containing SB medium. When the OD value at 600 nm reached about 0.5, IPTG was added to reach 1 mM, followed by incubation at 30° C. overnight, thereby expressing scFv proteins in the periplasm of E. coli. The next day, E. coli obtained through centrifugation were suspended in 1×TES buffer (50 mM Tris, 1 mM EDTA, 20% Sucrose, pH 8.0), and then 0.2×TES was added by 1.5 times, followed by mixing, and then supernatant was taken through centrifugation, thereby extracting the periplasm.

Finally, 5 mM MgSO4 was added to the scFv antibodies extracted from the periplasm, and the resultant material was mixed with Ni-NTA beads previously equilibrated with PBS, followed by stirring for 1 h in a cold storage to bind the antibody to the Ni-NTA beads. Thereafter, affinity chromatography was performed to sufficiently wash out the non-bound proteins with PBS. After further sufficient washing with a buffer containing 5 mM imidazole, the bound scFv antibodies were eluted using 200 mM imidazole buffer. The eluted antibodies were dialyzed, and the purity thereof was checked by electrophoresis. The protein quantification was performed by BCA assay, and the amount of purified antibodies was recorded, and then a certain amount thereof was dispensed and then frozen stored.

As described above, the scFv antibodies extracted from the periplasm were used to investigate using western blotting whether the scFv antibodies bound to the full-length KRS or respective KRS N-terminal fragments. Then, 30 μg of HCT116 cell lysate was electrophoresed through SDS PAGE, transferred onto PVDF membrane, and then blocked with 3% skim milk. Thereafter, the extracted scFv antibodies were added at 1.0 μg/μl, followed by incubation for 1 h. The unbound scFv antibodies were washed out, and for detection, the scFv binding with antigens were incubated with anti-HA secondary antibodies linked with horseradish peroxidase (HRP), and film sensitization was carried out using ECL reagent as a substrate in a dark room. The sensitized bands were compared with standard molecule markers to identify bands corresponding to sizes of the full-length KRS and the respective fragments.

Through the western blotting, scFv clones with highly weak bands (faint bands) and non-specific bands (double bands) were ruled out. Therefore, 13 scFv clones were selected, and these results are shown in FIG. 1.

<1-5> Screening of Specifically Binding scFv Antibodies Through Immunoprecipitation_Quaternary Screening

Immunoprecipitation was performed to investigate whether the scFv clones selected in Example <1-4> actually bound to native KRS. The purified scFv clones and HCT116 cell lysate were subjected to Ag-Ab binding, and immunoprecipitation was performed utilizing scFv HA-tag.

Specifically, HCT116 cells were lysed in 20 mM Tris-HCl buffer (pH 7.4, lysis buffer) containing 150 mM NaCl, 0.5% Triton X-100, 0.1% SDS and a protease inhibitor. Each scFv (5 μg) was added to 500 μg of HCT116 cell lysate, and then incubated at 4° C. overnight. Then 30 μl of anti-HA agarose beads were added, followed by incubation at 4° C. for 4 h. The supernatant was removed through centrifugation. The precipitate thus obtained was dissolved in SDS-sample buffer, and boiled for 7 min. The dissolving and boiling step was repeated twice.

Each of the immunoprecipitated samples prepared through the above-described procedure was electrophoresed through SDS PAGE, transferred onto PVDF membrane, and then blocked with 3% skim milk. Thereafter, KRS polyclonal antibodies (rabbit, Neomics, Co. Ltd. #NMS-01-0005) were added, followed by incubation for 1 h. After the unbound antibodies were washed out, anti-rabbit secondary antibodies (ThermoFisher Scientific, #31460) were added, followed by incubation. After incubation with the secondary antibodies, film sensitization was carried out using ECL reagent as a substrate in a dark room. The sensitized bands were compared with standard molecule markers to identify bands corresponding to sizes of the full-length KRS and the respective fragments.

Therefore, 12 scFv clones were selected, and these results are shown in FIG. 2.

<1-6> Screening of Specifically Binding scFv Antibodies Through Immunofluorescence_Quinary Screening

Immunofluorescence was performed to investigate whether the scFv clones selected in Example <1-5> actually bound to the KRS N-terminal region exposed on the cell membrane. In order to make a phenomenon in which KRS is exposed on the membrane, KRS-T52D mutant (active mutant) was used. Specifically, A549 cells were seeded on glass coverslip (1×105 cell, 12 well plate-based), and after 24 h, myc-KRS WT/myc-KRS T52D (active mutant)/myc-KRS T52A (inactive mutant) were overexpressed, respectively. After incubation for 24 h, the cells were incubated using serum-free media (RPMI 1640 media) at 37° C. for 1 h. Thereafter, the cells were treated with 10 μg/ml laminin (L2020; Sigma) and then incubated at 37° C. for 1 h. Each of myc-KRS T52D (active mutant) and myc-KRS T52A (inactive mutant) vectors was constructed using pcDNA3-myc-KRS WT vector as a backbone through site-directed mutagenesis (QuikChange □ Site-Directed Mutagenesis kit, Agilent, #200523).

The prepared samples were washed with PBS (4° C.), fixed by the treatment with 4% paraformaldehyde for 10 min, and then washed. The samples were blocked with CAS block for 10 min, and treated with the scFv and Myc antibodies for 2 h. Thereafter, the unbound antibodies were washed out, and incubated with secondary antibodies for 1 h in a dark room. DAPI staining was performed for 10 min before mounting.

As shown in FIGS. 3A-3C, the experimental results showed that a total of four scFv clones (N3, N5, N7, and N9) not binding to the inactive mutant (T52A) and WT-KRS (laminin untreated) but binding only active mutant cells were selected. When laminin treatment was carried out for inducing the membrane localization of WT-KRS and scFv staining was performed, similar staining regions were observed in WT-KRS and T52D mutant.

It was confirmed that these clones specifically bind to the cell surface (tip).

<1-7> Verification of Specific Binding Ability to KRS N-Terminus

To investigate whether the scFv clones (N3, N5, N7, and N9) finally selected through Example <1-6> actually bound to KRS N-terminus, full-length KRS (denoted by F, SEQ ID NO: 76), a KRS fragment with deletion in amino acids at positions 1-71 in the N-terminus (defined by SEQ ID NO: 1), and a KRS fragment composed of amino acid residues 1-200 in the N-terminus (defined by SEQ ID NO: 2) were used to conduct western blotting.

A549 cells were transformed by the method as described in Example <1-6> using polynucleotides encoding full-length KRS and KRS fragments. Thereafter, the cells were lysed, and western blotting was performed by the same method as described in Example <1-4>.

As shown in FIG. 4, the results confirmed that all the selected scFv clones (N3, N5, N7, and N9) showed bands in only the fragment with amino acids at 1-200 in the N-terminus while showing no bands in fragment 1 without amino acids at positions 1-72 in the N-terminus. It was therefore verified that all of the scFv N3, N5, N7, and N9 clones specifically bound to the KRS N-terminus.

<1-8> Sequencing of KRS N-Terminal Binding Specific scFv Clones

The scFv clones (N3, N5, N7, and N9) finally selected through Example <1-6> were analyzed for CDR conformation and VH and VL sequences thereof. Sequencing was performed by the same method as described in Example <1-3>.

As a result of sequencing, N3 scFv consists of the amino acid sequence defined by SEQ ID NO: 67, which contains the linker sequence of SEQ ID NO: 65 in the middle thereof. In addition, N3 VH consists of the amino acid sequence defined by SEQ ID NO: 49 and N3 VL consists of the amino acid sequence defined by SEQ ID NO: 51. As a result of sequencing respective CDRs contained in VH and VL of N3, N3 VH comprises heavy chain CDR1 defined by SEQ ID NO: 1, heavy chain CDR2 defined by SEQ ID NO: 3, and heavy chain CDR3 defined by SEQ ID NO: 5, and N3 VL comprises light chain CDR1 defined by SEQ ID NO: 7, light chain CDR2 defined by SEQ ID NO: 9, and light chain CDR3 defined by SEQ ID NO: 11.

N5 scFv consists of the amino acid sequence defined by SEQ ID NO: 69, which contains the linker sequence of SEQ ID NO: 65 in the middle thereof. In addition, N5 VH consists of the amino acid sequence defined by SEQ ID NO: 53 and N5 VL consists of the amino acid sequence defined by SEQ ID NO: 55. N5 VH comprises heavy chain CDR1 defined by SEQ ID NO: 13, heavy chain CDR2 defined by SEQ ID NO: 15, and heavy chain CDR3 defined by SEQ ID NO: 17, and N5 VL comprises light chain CDR1 defined by SEQ ID NO: 19, light chain CDR2 defined by SEQ ID NO: 21, and light chain CDR3 defined by SEQ ID NO: 23.

N7 scFv consists of the amino acid sequence defined by SEQ ID NO: 71, which contains the linker sequence of SEQ ID NO: 65 in the middle thereof. In addition, N7 VH consists of the amino acid sequence defined by SEQ ID NO: 57 and N7 VL consists of the amino acid sequence defined by SEQ ID NO: 59. N7 VH comprises heavy chain CDR1 defined by SEQ ID NO: 25, heavy chain CDR2 defined by SEQ ID NO: 27, and heavy chain CDR3 defined by SEQ ID NO: 29, and N7 VL comprises light chain CDR1 defined by SEQ ID NO: 31, light chain CDR2 defined by SEQ ID NO: 33, and light chain CDR3 defined by SEQ ID NO: 35.

N9 scFv consists of the amino acid sequence defined by SEQ ID NO: 73, which contains the linker sequence of SEQ ID NO: 65 in the middle thereof. In addition, N9 VH consists of the amino acid sequence defined by SEQ ID NO: 61 and N9 VL consists of the amino acid sequence defined by SEQ ID NO: 63. N9 VH comprises heavy chain CDR1 defined by SEQ ID NO: 37, heavy chain CDR2 defined by SEQ ID NO: 39, and heavy chain CDR3 defined by SEQ ID NO: 41, and N9 VL comprises light chain CDR1 defined by SEQ ID NO: 43, light chain CDR2 defined by SEQ ID NO: 45, and light chain CDR3 defined by SEQ ID NO: 47.

Example 2

Conversion of scFv Antibodies into IgG Antibodies and Evaluation of Specific Binding Ability Thereof

<2-1> Conversion of scFv Antibodies into IgG Antibodies

First, the polynucleotides encoding scFv were amplified via PCR from N3, N5, N7, and N9 phage genomes. The nucleotide sequences of the primers used to amplify a gene of VH region of the scFv antibodies: Forward (AGA GAG TGT ACA CTC CCA GGC GGC CGA GGT GCA G, SEQ ID NO: 93), Reverse (CGC CGC TGG GCC CTT GGT GGA GGC TGA GCT CAC GGT GAC CAG, SEQ ID NO: 94). The nucleotide sequences of the primers used to amplify a gene of VL region of the scFv antibodies: Forward (AAG CGG CCG CCA CCA TGG GAT GGA GCT GTA TCA TCC TCT TCT TGG TAG CAA CAG CTA CAG GTG TAC ACT CCC AGT CTG TGC TGA CTC AG, SEQ ID NO: 95), Reverse (CGC CGC CGT ACG TAG GAC CGT CAG CTT GGT, SEQ ID NO: 96)

PCR was performed with each phage DNA (50 ng) as a template by using the primers (10 pmol each) in conditions of: 95° C./3 min; 95° C./30 s, 60° C./30 s, 72° C./30 s, 30 cycles; and 72° C./5 min, thereby amplifying the VH or VL gene of N3, N5, N7, or N9 scFv. The PCR product was inserted into the pcDNA3.4 vector used in IgG production using restriction enzymes. IgG heavy and light chain proteins were individually encoded in separate plasmids.

The constructed vectors comprising DNA encoding heavy and light chains of each of IgGs (hereinafter, called N3 IgG, N5 IgG, N7 IgG, and N9 IgG, respectively) containing scFv variable regions were co-transformed in freestyle 293F cells to express the heavy and light chains together in cells. The transformed 293F cells were incubated in conditions of 37° C. and 8% CO2 for 7 days, and the supernatant was obtained. The supernatant was filtered through a cellulose acetate membrane filter (pore size 0.22 μm, Corning), and purified using CaptivA™ PriMAB protein A column (Repligen, USA). The concentrations of the obtained antibodies were measured using BCA kit (Pierce, 23225), and the IgG antibody proteins produced in reduction and non-reduction conditions were analyzed.

<2-2> Verification of KRS Binding Ability of Converted IgG_Western Blotting and Immunoprecipitation

The KRS binding ability of the IgGs constructed in Example <2-1> was investigated by western blotting (WB) and immunoprecipitation. Western blotting was performed in the same manner as described in Example <1-4> and immunoprecipitation was performed in the same manner as described in Example <1-5>.

The results verified that the IgGs constructed in the present invention bound to KRS, and FIGS. 5A-5B shows these results using N3 IgG and N5 IgG as representatives.

<2-3> Verification of KRS N-Terminus-Specific Binding Ability of Converted IgGs_SPR

The quantitative binding ability of the purified antibody proteins (N3 IgG and N5 IgG) to the antigen (KRS 1-207 aa) were measured using Biacore 2000 SPR (surface plasmon resonance) (GE healthcare, US) biosensor. After KRS was immobilized on a sensor chip (CM5, GE healthcare, US), antibody proteins (6.25-100 nM), which were serially diluted with HES buffer solution (10 mM HEPES, pH 7.4, 150 mM NaCl, 3 mM EDTA, 0.005% surfactant P20), were allowed to flow at a rate of 30 μl/min for 3 min, and 1 M NaCl/20 mM NaOH was allowed to flow at a rate of 30 μl/min for 3 min, thereby inducing the dissociation of proteins bound to the antigen. Mock IgG was used as control. Specific experiment conditions are as follows:

Immobilized Antigen: KRS

Immobilized level: 185 RU

Antibody: N3 IgG and N5 IgG

Running buffer: HBS-N buffer

Regeneration: 2 M NaCl, 20 mM NaOH (flow 30 ul/min 1 min)

TABLE 3 ka (1/Ms) kd (1/s) KD (M) N3 IgG 1.09E+05 0.009055 8.34E−08 N5 IgG 3.13E+06 0.003282 1.05E−09

Table 3 shows kinetic rate constants and equilibrium dissociation constants measured for N3 IgG and N5 IgG by using Biacore 2000 SPR. The affinity was obtained from the kinetic rate constants (ka and kd) and equilibrium dissociation constants (KD) by using BIA evaluation ver. 3.2 software. FIGS. 6A-6B show SPR graph results of N3 IgG and N5 IgG, respectively. It was confirmed from FIGS. 6A-6B and Table 3 that N3 IgG and N5 IgG of the present invention has high specific binding ability to the KRS N-terminal region. No binding signals were observed in mock IgG as control.

<2-4> Verification of KRS N-Terminus Specific Binding Ability of Converted IgG Immunofluorescence Staining

To investigate whether the IgGs constructed in the present invention actually bound to the region of KRS exposed on the cell membrane, immunofluorescence was performed. Immunofluorescence staining was performed by the same method as described in Example <1-6> by using N3 IgG and N5 IgG as representatives.

As shown in FIG. 7A, the results confirmed that N3 IgG favorably bound specifically to the extracellularly exposed KRS N-terminal region when the membrane localization of WT-KRS was induced by laminin treatment. As shown in FIG. 7B, the experiment using T52D-KRS (active mutant, myc tagged KRS) also showed that the antibody of the present invention favorably bound to the extracellularly exposed KRS N-terminal region, and when the experimental cells became permeabilized, the KRS proteins present in cytosol were detected at high sensitivity. It was also confirmed as shown in FIG. 7C that N5 IgG favorably bound specifically to extracellularly exposed KRS N-terminal region when the membrane localization of WT-KRS was induced by laminin treatment.

It was therefore verified that the antibodies provided in the present invention have high binding specificity to an extracellularly exposed KRS N-terminal region.

Example 3

Verification of Inhibitory Effect on Cancer Metastasis

<3-1> Cytotoxicity Evaluation

MTT assay was performed on N3 IgG as a representative. A549 cells were seeded in 96-well plates (5,000 cell/well) and incubated. The cells were washed with serum-free media, and then treated with human mock IgG and N3 IgG of 0, 50, 100, 500 nM (in serum-free media). After 24-h incubation, MTT solution was added with 50 μg/well, followed by incubation for 4 h. After MTT solution was removed, the wells were treated with 100 ul of DMSO, and then the absorbance was measured at 570 nm.

As shown in FIG. 8, the experimental results confirmed that the antibody of the present invention shows no cytotoxicity.

<3-2> Cell Migration Assay

Cell migration was measured using a 24-well Transwell chamber with polycarbonate membrane (8.0 μm pore size, Costar) as disclosed in the prior art (Park, S. G. et al., Human lysyl-tRNA synthetase is secreted to trigger pro-inflammatory response, Proc. Natl. Acad. Sci. USA 102, 6356-6361 (2005)). In the Transwell chamber, the lower well was coated with 10 μg of laminin (in gelatin) and dried with UV. Thereafter, A549 cells were suspended in serum-free RPIM media, and then placed at a concentration of 1×105 cells per well in the upper chamber. The chamber was treated with N3 IgG or human mock IgG (control) at 100 nM or 500 nM, followed by incubation for 24 h. Thereafter, the chamber was washed twice with PBS, and treated with 70% MeOH (in PBS) for 30 min. The chamber was again washed twice with PBS, and treated with Hematoxylin solution for 30 min. The chamber was washed three times with DW, and the membrane in the chamber was cut and mounted on the slide glass.

As shown in FIG. 9A, the experimental results showed that N3 IgG significantly inhibited the migration of A549 cells. The experimental results also showed that this cell migration inhibitory effect was dose-dependent (see FIG. 9B).

<3-3> Evaluation of Cancer Metastasis Inhibitory Effect in In-Vivo Cancer Metastasis Models

Since KRS can accelerate cell migration through 67LR associated with cancer metastasis, tumor (cancer) animal models were constructed using mouse breast cancer 4T-1 cells (Korean Cell Line Bank), which are well metastasizable to the lung. Orthotopic breast cancer animal models were constructed by injecting 4×104 4 T1 cells into the fat pad of six 7-week old BALB/cAnCr mice (Doo Yeol Biotech).

The cancer was injected into the mammary fat pad, and after 10 days (Day 10), cancer tissues were resected from the fat pad. After one day (Day 11), N3 IgG (10 mg/kg) was administered twice a week for two weeks at intervals of 3 days (a total of four times, Days 11, 14, 18, and 21) via tail vein i.v. injection, and the same dose of control mock IgG (Thermo #31154) was also administered. One week after the completion of the entire antibody dosing schedule, that is, 28 days after cancer injection, the mice were sacrificed to take lung tissue. Upon autopsy of lung tissue, the lung was inflated by injection of a saline solution into the bronchus through a syringe, collected, and then stored in Bouin's solution (Sigma #HT10132) for 24 h. Thereafter, the metastasis nodules in each lobe of the lung were counted through a microscope.

As shown in FIGS. 11 and 12, the experimental results confirmed that many nodules were generated in the lung due to cancer metastasis in control, and such nodules were significantly suppressed in the N3 IgG treatment group. FIG. 13 comparatively shows the lung tissues in control and the N3 IgG treatment group, and confirmed that a significantly large amount of laminin receptors were expressed in the metastasis nodule sites in control compared with the group treated with the antibody of the present invention.

<3-4> Effect Comparison with Anti-Cancer Metastasis Compound (YH16899) in In-Vivo Cancer Metastasis Models

It has been known in the foregoing literature “Dae Gyu Kim et al., (2014)” that the YH16899 compound has an effect on cancer metastasis inhibition by suppressing the interaction between 67LR and KRS. Then, the cancer metastasis inhibitory effect was compared between YH16899 and N3 IgG, the antibody of the present invention. The construction of in-vivo tumor models and the observation of lung metastasis condition were carried out by the same method as in Example <3-3>. YH16899 was orally administered at 100 mpk every day. N3 IgG was intravenously injected at difference concentrations (1 mpk, 10 mpk) through mouse tails.

As shown in FIGS. 14 and 15, the experimental results confirmed that the lung nodule count was significantly reduced in a dose-dependent manner in the N3 IgG treatment group, and the treatment with merely 1 mpk of N3 IgG significantly inhibited cancer metastasis compared with YH16899 (100 mpk) treatment groups.

Example 4

Verification of Binding Sites of KRS Antibodies

<4-1> Human KRS Binding Sites of KRS Monoclonal Antibodies

To investigate human KRS binding sites of N3 IgG among the KRS antibodies constructed above, surface plasmon resonance (SPR) was performed as below.

First, N3 IgG antibody was immobilized to Biacore T200 (GE Healthcare) equipped with Series S sensor chip CM5 (GE Healthcare) by using an amine coupling kit (GE Healthcare). Then, the peptides shown in Table 4 below dissolved in PBS solution at corresponding concentrations were allowed to flow for 60 s. Then, PBS was allowed to flow for 5 min. Then, the binding ability was analyzed by Biacore T200 Evaluation software v2.0 (GE Healthcare).

TABLE 4  Peptide informaiton SEQ ID Name Sequence information Species MW NO F1 (1-29) MAAVQAAEVKVDGSEPKLSE H 3168 98 ELKRPIKA F2 (5-34) QAAEVKVDGSEPKLSEELKR H 3351 99 PIKEKKVA F3 (10-38) KVDGSEPELSENELERRLKA H 3310 100 EKKVAEKEA F4 (15-42) EPKLSKNELKRRLKAEKKVA H 3337 101 EKEAKQKE F5 (24-49) KRRLKAEKKVAEKEAKQKEL H 3084 102 SEKQLS mF1 (1-28) MAILQESEVEVDGEQKLSKN M 3230 103 ELKRRLKA mF2 (3-34) QESEVEVDGEQKLSKNELKR M 3383 104 RLKAEKKLA mF3 (10-37) KVDGEQKLSKNELKPRLKAE M 3268 105 KKLAEKEA mF4 (15-41) QELSENELERRLKAEKKLAE M 3253 106 KEAKQEE mF5 (24-48) RRLKAEKELAEKEAKQKELS M 2997 107 EKQLN rF1 (1-28) MAILREGEVELDGEPKLSKN R 3211 108 ELKRRLKA rF2 (3-34) REGEVELDGEPKLSKNELKR R 3364 109 RLKAEKKLA rF3 (10-37) KLDGEPKLSKNELKRRLKAE R 3251 110 KKLAEKEA rF4 (15-41) PKLSKNELKRRLKAEKKLAE R 3222 111 KEAKQKE rF5 (24-48) RRLKAEKKLAEKEAKQKELS R 2997 112 EKQLN

As shown in FIG. 16, the results depicted that N3 IgG antibody bound to epitopes F1, F2, F3, and F4, but not epitope F5. In addition, the binding ability to epitope F4 was strongest, and the binding ability to F3, F2, and F1 was stronger in that order.

These results could confirm that the main binding site of N3 IgG antibody corresponds to amino acid resides at positions 15 to 29 in the KRS N-terminal region.

<4-2> Interspecies Cross Activity of KRS Monoclonal Antibody

The above example validated the human KRS binding site of the KRS antibody N3 IgG, and to investigate whether N3 IgG showed cross activity with other species mouse (m) and rat (r), surface plasmon resonance (SPR) was performed as below.

In the same manner as the experimental method described in Example 4-1 above, N3 IgG antibody was immobilized on the chip by using an amine coupling kit (GE Healthcare). Then, the peptides shown in Table 4 above dissolved in PBS solution at corresponding concentrations were allowed to flow for 60 s, and PBS was allowed to flow for 5 min. Then, the binding ability was analyzed by Biacore T200 Evaluation software v2.0 (GE Healthcare).

As shown in FIG. 17, the results depicted that the N3 IgG antibody bound to epitopes F1, F2, F3, and F4 of human (h), mouse (m), and rat (r) but not epitope F5. The stronger binding ability to epitope F3 than F1 was the same among human, mouse, and rat (F2 and F4 data not shown)

These results could confirm that the N3 IgG antibody is capable of interspecies cross activity.

Example 5

Verification of Laminin Signal Role in Immune Cell Migration and Invasion

It was investigated which of several extracellular matrixes constituting blood vessels accelerated the migration and invasion of monocytes/macrophages. Specific experimental methods for Transwell migration assay using collagen, fibronectin, and laminin as extracellular matrixes were as below. Transwell (Corning, #3421-5 mm) was coated with gelatin (0.5 mg/ml), and then RAW 264.7 cells (1×105 cells/well) were seeded in the top chamber. Each serum-free DMEM (500 μl) containing laminin (laminin mixture, Biolamina), fibronectin, or collagen (10 μg/ml) was placed in the bottom chamber. After 24 h, non-migrating cells present on the upper part of the membrane were removed by cotton swabs. The cells in the bottom chamber were fixed by treatment with 70% methanol for 30 min, and then stained with 50% hematoxylin for 30 min. After the staining, the membrane was taken and mounted on the slide, and then the migrating cells present on the bottom surface of the membrane were observed and quantified through a high-resolution microscope.

As shown in FIGS. 18A and 18B, the experimental results confirmed that out of several extracellular matrixes, laminin accelerated monocyte/macrophage migration most strongly.

Example 6

Immune Cell Migration and Invasion Effects by Laminin Subtypes

The effects of laminin subtypes on the immune cell migration and invasion were evaluated. Transwell migration assay was performed by the same method as in Example 5 using LN111, LN211, LN221, LN411, LN421, LN511, and LN521 (10 μg/ml) as various laminin subtype proteins (purchased from Biolamina). Specific sequences of the laminin subtypes may be referenced α4 chain of SEQ ID NO: 115, α2 chain of SEQ ID NO: 121, α5 chain of SEQ ID NO: 122, β2 chain of SEQ ID NO: 117, β1 chain of SEQ ID NO: 123, and γ1 chain of SEQ ID NO: 119, according to the chains constituting respective laminin subtypes.

RAW 264.7 cells (2×106 cell) were incubated for 18 hr, treated with 1 μg/ml of each laminin subtype in serum-free DMEM, and then harvested at 0 h, 12 h, and 24 h. RAW 264.7 cell protein was separated into cytosol and membrane fractions by using the ProteoExtract Subcellular Proteome Extraction Kit (Calbiotech, cat #539790). The obtained protein was electrophoresed, transferred onto PVDF membrane (Milipore), and blocked with 3% skim milk. Thereafter, KRS polyclonal antibody (rabbit, Neomics, Co. Ltd. #NMS-01-0005) was added, followed by incubation for 1 h. After the unbound antibodies were washed out, anti-rabbit secondary antibodies (ThermoFisher Scientific, #31460) were added, followed by incubation. After incubation with the secondary antibodies, film sensitization was carried out using ECL reagent as a substrate in a dark room. The sensitized bands were compared with standard molecule markers to identify bands corresponding to KRS sizes. Na+/K+ ATPase (Abcam) and tubulin (Sigma) antibody were used for plasma membrane and cytosol marker identification, respectively.

As shown in FIGS. 19A and 19B, the experiment results confirmed that monocytes/macrophages migrate by specifically responding to α4β2γ1 subtype (LN421) among all the tested laminin subtypes. That is, it was confirmed that monocytes/macrophages migrated and invaded specifically responding to LN421. As shown in FIG. 19C, it was confirmed that the treatment of monocytes/macrophages with LN421 increased the amount of KRS detected in the cellular membrane region but partly decreased the amount of KRS detected in the cytosol region. These results indicate that KRS, which is generally present in the cytosol region after expression inside monocytes/macrophages, translocates to the cellular membrane region by LN421 treatment, and that a KRS increase in the immune cell membrane region corresponds to an important pathological phenomenon in the diseases associated with immune cell migration and invasion.

Example 7

Construction of Antibody for Reducing Cellular Membrane KRS Level and Verification of Immune Cell Migration/Invasion Control Effect

The effect on immune cell migration and invasion was investigated using N3 IgG antibody as a representative among the antibodies constructed in Example 1 above. Specific experimental methods were as follows. Transwell (Corning, #3421-5 mm) was coated with gelatin (0.5 mg/ml), and then RAW 264.7 cells (1×105 cells/well) were seeded in the top chamber. Serum-free DMEM (500 μl) containing laminin 421 (1 μg/ml) was placed in the bottom chamber. The top chamber was treated with each antibody at 100 nM. After 24 h, immobilization with 70% methanol was carried out for 30 min, and then staining with 50% hematoxylin was carried out for 30 min. The non-migrating cells present on the upper part of the membrane were removed by cotton swabs, and then the membrane was taken and mounted on the slide. The migrating cells present on the bottom surface of the membrane were observed by a high-resolution microscope (FIG. 20A), and the cells were counted on the obtained images and plotted on the graph (FIG. 20B).

RAW 264.7 cells were treated with laminin 421 (1 μg/ml) and antibody (100 nM), incubated for 24 h, and harvested. Thereafter, the harvest was separated into the membrane and cytosol fractions by using the ProteoExtract Subcellular Proteome Extraction Kit (Calbiochem), sampled, and then subjected to western blotting with respect to KRS. Specific method thereof was as described in Example 6.

As shown in FIGS. 20A and 20B, the experimental results confirmed that the antibody of the present invention effectively inhibited specifically LN421-dependent monocyte/macrophage migration. As shown in FIG. 20c, it was confirmed that the LN421 treatment increased the KRS level in the monocyte/macrophage cell membrane and the treatment with N3 IgG antibody effectively reduced the KRS level in the cell membrane.

These results confirmed that the antibody of the present invention has possibility as a novel therapeutic agent for diseases involved in the migration of immune cells, such as monocytes/macrophages.

Example 8

Verification of Effect on Immune Cell Migration-Related Disease in In-Vivo Models

As in the examples above, the following experiment was executed using N3 IgG antibody as a representative among the antibodies of the present invention.

[Methods]

1. Pulmonary Arterial Hypertension (PAH) Model Construction and Test Substance Administration

To induce PAH in seven-week-old SD rats (Orient Bio), 60 mpk of monocrotaline (MCT) was subcutaneously injected. Thereafter, the rats were divided into four groups (five animals per group), and administered with 1 mpk of mock human IgG (Thermo Fisher Scientific, negative control), 1 mpk of N3 IgG, 10 mpk of N3 IgG 10, and 25 mpk of sildenafil (positive control) for three weeks. All antibodies were i.v. injected twice a week, and sildenafil was orally administered every day.

2. Blood Flow and Blood Pressure Measurement

After three weeks, the rats were anesthetized with isoflurane, and measured for blood flow and pressure by using a high-precision pneumatic measurement system (MPVS cardiovascular pressure and volume system, model name: MPVS Ultra, manufacturer: Millar Instruments). The right ventricular end-systolic pressure (RVESP), right ventricular end-diastolic pressure, left ventricular end-systolic pressure, left ventricular end-diastolic pressure were measured using an exclusive catheter (Mikro-Tip rat pressure catheter, manufacturer: Millar Instruments). The cardiac output was measured using a perivascular blood flow probe (Transonic Flow probes, manufacturer: Millar Instruments), and experimental method thereof was performed by the same method as disclosed in the following literature: Pacher P, Nagayama T, Mukhopadhyay P, Batkai S, Kass D A. Measurement of cardiac function using pressure-volume conductance catheter technique in mice and rats. Nat Protoc 2008; 3(9):1422-34.

3. Immunohistochemistry (IHC)

The collected lungs were fixed in paraformaldehyde (PFA) according to the ordinary procedure, and then paraffin-infiltrated and embedded through washing, dehydration, and clearing. The rat lung tissue paraffin blocks were micro-sectioned to a thickness of 6 μm, and slides were manufactured. Thereafter, staining was performed as below. The sample was first treated with xylene for 5 min three times, treated with 100% ethanol, 95% ethanol, 90% ethanol, and 70% ethanol, and DW in that order for 2 min, and washed with PBS for 5 min. After 0.3% H2O2 treatment, the sample was washed with PBS for 5 min twice. The sample was immersed in 0.01 M citrate buffer, heated, and washed with PBS-T (0.03% Tween 20). Thereafter, the sample was blocked (2% BSA & 2% goat serum in PBS) at room temperature for 30 min. The sample was stained with anti-CD68 antibody (1:200, ED1 clone, Abcam) at 4° C. overnight. The sample was washed with PBS-T for 5 min three times, and then treated with polymer-HRP anti-mouse envision kit (DAKO) at 4° C. for 1 h. The sample was washed with PBS-T three times, and then color-developed by the treatment with DAB substrate buffer and DAB chromogen 20. The stained tissue was treated with Mayer's hematoxylin (Sigma) for 1 min, and then treated with 70% ethanol, 90% ethanol, 95% ethanol, and 100% ethanol in that order for 2 min each twice. Last, the tissue was treated with xylene three times for 5 min, and then observed under an optical microscope.

[Results]

<2-1> Verification of Blood Pressure and Cardiac Output Changes.

The models of PAH, which is a disease having a close relation between immune cell invasion and pathological phenomena, were treated with N3 IgG antibody (1 mpk or 10 mpk) for 3 weeks (i.v., twice a week), and then measured for right ventricular end-systolic pressure (RVESP), right ventricular end-diastolic pressure (RVEDP), left ventricular end-systolic pressure (LVESP), left ventricular end-diastolic pressure (LVEDP), and cardiac output (CO). The results thereof are shown in Table 5.

TABLE 5 MCT + MCT + MCT + N3 Ab N3 Ab MCT + Mock IgG 1 mpk 10 mpk Sildenafil (n = 4) (n = 5) (n = 5) (n = 5) RVESP 62.5 ± 5.7  45.0 ± 8.1 41.2 ± 7.7  48.4 ± 9.6  (mmHg) RVEDP 2.8 ± 1.5  1.4 ± 2.2 3.8 ± 1.3 2.6 ± 1.3 (mmHg) LVESP 81.5 ± 11.4 95.8 ± 4.8 93.4 ± 11.3 83.2 ± 4.7  (mmHg) LVEDP 1.0 ± 0.8  2.6 ± 1.9 4.6 ± 3.9 3.6 ± 2.3 (mmHg) CO  58 ± 4.7 74.0 ± 0.9 59.8 ± 12.9 49.6 ± 17.7 (ml/min) (n = 4) (n = 5) (n = 5) (n = 4) (No CO measurement for one animal of MCT + mock IgG group, died from anesthesia, and one animal of sildenafil treatment group, died during surgery)

Pulmonary arterial hypertension causes the right ventricular-end pressure to rise due to narrowing of the pulmonary artery, resulting in right ventricular failure. In addition, the reward mechanism thereof is destroyed due to continuous hypertension, resulting in right ventricular hypertrophy followed by right ventricular dilation. This causes left ventricular compression due to ventricular septum movement, resulting in reductions in end-diastolic volume and cardiac output of the left ventricle (WooSeok Lee, et al., Clinical Characteristics and Prognostic Factors of Patients with Severe Pulmonary Hypertension, Korean Circulation J 2007; 37:265-270). Resultingly, the pulmonary arterial hypertension is mainly associated with the right ventricle, but also involved in functions of the left ventricle.

PAH patients showed a RVESP increase, which was also observed in the PAH animal models of the present experiment. In this regard, as shown in FIG. 21A, N3 antibody significantly reduced RVESP at both the concentrations thereof, and favorably reduced RVESP than especially sildenafil, the positive control drug.

In addition, a reduction in left ventricular end-systolic pressure (LVESP) due to N3 IgG antibody administration was not observed, and rather, as shown in FIG. 21B, LVESP significantly increased in the group administered with the antibody of the present invention. Therefore, the antibody of the present invention is contrast with sildenafil used as an existing therapeutic agent for pulmonary arterial hypertension wherein sildenafil causes pulmonary arterial dilatation and systemic arterial dilatation, thereby risking a reduction in systemic blood pressure.

That is, it was confirmed that the antibody of the present invention showed a tendency of having a low effect on systemic artery pressure compared with sildenafil, and this effect is thought to be a favorable characteristic of a therapeutic agent considering that sildenafil administration may be a risk of developing hypotension in clinical sites. Moreover, severe pulmonary arterial hypertension causes systolic RV failure, which may be accompanied by low cardiac output and systemic hypotension.

Whereas, a treatment to alleviate pulmonary arterial hypertension by the antibody of the present invention is expected to increase the cardiac output and systemic blood pressure, thereby normalizing the blood pressure.

Overall, it was confirmed that the administration of the antibody of the present invention reduced the risk of side effects of existing therapeutic drugs and showed PAH symptom alleviation and treatment effects.

<8-2> Echocardiography

The D-shaped left ventricle finding indicating pressure overload in the right ventricle was observed in three animals in the MCT alone administration group (i.e., test substance non-administration PAH models) and three animals in the MCT+sildenafil administration group, but was not observed in the therapeutic antibody administration groups.

In addition, as shown in Table 6 below, the body weights of respective groups increased to similar degrees, with no significant difference. That is, the findings were not observed to indicate abnormal signs, including abnormal weight reduction, caused by the administration of the therapeutic antibody.

TABLE 6 MCT + Mock MCT + Ab MCT + Ab MCT + IgG 1 mpk 10 mpk Sildenafil (n = 4) (n = 5) (n = 5) (n = 5) Absolute 101.4 ± 14.2 113.5 ± 14.6 104.1 ± 12.3 104.1 ± 26.4 change (g) Relative 48.8 ± 7.8 43.6 ± 5.2 40.7 ± 5.0  49.8 ± 10.5 change (%)

<8-3> Verification of Monocyte/Macrophage Migration and Infiltration Degrees

IHC staining was performed with respect to CD68, which is a monocyte/macrophage marker, by using the lung tissues of each experimental group. As shown in FIG. 22, the experimental results confirmed that the groups treated with N3 IgG antibodies of the present invention explicitly reduced the monocyte/macrophage infiltration into lung tissues, and such an effect was significantly excellent than that of sildenafil.

INDUSTRIAL APPLICABILITY

As described above, the antibodies or fragments thereof according to the present invention have particular CDR (complementary determining region) sequences defined in the present specification and very excellent specific binding ability to the extracellularly exposed KRS N-terminal region, and thus can be used in the diagnosis of a disease (e.g., cancer) known to be accompanied by specific behaviors of KRS. Furthermore, the antibodies or fragments thereof according to the present invention are also specifically targeted to the KRS N-terminal region in vivo, and thus inhibit the interaction between the laminin receptor and the KRS N-terminal region to exert an excellent inhibitory effect on cancer metastasis, and therefore can be used as a therapeutic agent. Furthermore, the antibodies or fragments thereof according to the present invention can control the migration of immune cells, and thus can be very advantageously used in the prevention, alleviation, and treatment of immune cell migration-related diseases, and therefore have high industrial applicability.

Claims

1. An antibody or fragment thereof specifically binding to an epitope consisting of a sequence of SEQ ID NO: 101, SEQ ID NO: 106, or SEQ ID NO: 111 in the lysyl-tRNA synthetase (KRS) N-terminus, wherein the antibody or fragment thereof specifically binds to an extracellularly exposed lysyl-tRNA synthetase (KRS) N-terminal region, wherein the antibody or fragment thereof comprises a heavy chain variable region and a light chain variable region which are selected from the group consisting of:

a heavy chain variable region comprising heavy chain complementary determining region 1 containing the amino acid sequence defined by SEQ ID NO: 1, heavy chain complementary determining region 2 containing the amino acid sequence defined by SEQ ID NO: 3, and heavy chain complementary determining region 3 containing the amino acid sequence defined by SEQ ID NO: 5, and a light chain variable region comprising light chain complementary determining region 1 containing the amino acid sequence defined by SEQ ID NO: 7, light chain complementary determining region 2 containing the amino acid sequence defined by SEQ ID NO: 9, and light chain complementary determining region 3 containing the amino acid sequence defined by SEQ ID NO: 11;
a heavy chain variable region comprising heavy chain complementary determining region 1 containing the amino acid sequence defined by SEQ ID NO: 13, heavy chain complementary determining region 2 containing the amino acid sequence defined by SEQ ID NO: 15, and heavy chain complementary determining region 3 containing the amino acid sequence defined by SEQ ID NO: 17, and a light chain variable region comprising light chain complementary determining region 1 containing the amino acid sequence defined by SEQ ID NO: 19, light chain complementary determining region 2 containing the amino acid sequence defined by SEQ ID NO: 21, and light chain complementary determining region 3 containing the amino acid sequence defined by SEQ ID NO: 23;
a heavy chain variable region comprising heavy chain complementary determining region 1 containing the amino acid sequence defined by SEQ ID NO: 25, heavy chain complementary determining region 2 containing the amino acid sequence defined by SEQ ID NO: 27, and heavy chain complementary determining region 3 containing the amino acid sequence defined by SEQ ID NO: 29, and a light chain variable region comprising light chain complementary determining region 1 containing the amino acid sequence defined by SEQ ID NO: 31, light chain complementary determining region 2 containing the amino acid sequence defined by SEQ ID NO: 33, and light chain complementary determining region 3 containing the amino acid sequence defined by SEQ ID NO: 35; and
a heavy chain variable region comprising heavy chain complementary determining region 1 containing the amino acid sequence defined by SEQ ID NO: 37, heavy chain complementary determining region 2 containing the amino acid sequence defined by SEQ ID NO: 39, and heavy chain complementary determining region 3 containing the amino acid sequence defined by SEQ ID NO: 41, and a light chain variable region comprising light chain complementary determining region 1 containing the amino acid sequence defined by SEQ ID NO: 43, light chain complementary determining region 2 containing the amino acid sequence defined by SEQ ID NO: 45, and light chain complementary determining region 3 containing the amino acid sequence defined by SEQ ID NO: 47.

2. The antibody or fragment thereof of claim 1, wherein the antibody or fragment thereof comprises the heavy chain variable region containing the amino acid sequence selected from the group consisting of SEQ ID NO: 49, SEQ ID NO: 53, SEQ ID NO: 57, and SEQ ID NO: 61, and the light chain variable region containing the amino acid sequence selected from the group consisting of SEQ ID NO: 51, SEQ ID NO: 55, SEQ ID NO: 59, and SEQ ID NO: 63.

3. The antibody or fragment thereof of claim 1, wherein the antibody is selected from the group consisting of IgG, IgA, IgM, IgE, and IgD, and the fragment is selected from the group consisting of diabody, Fab, Fab′, F(ab)2, F(ab′)2, Fv, and scFv.

4. The antibody or fragment thereof of claim 3, wherein the scFv contains the amino acid sequence selected from the group consisting of SEQ ID NO: 67, SEQ ID NO: 69, SEQ ID NO: 71, and SEQ ID NO: 73.

5. A polynucleotide encoding the antibody or fragment thereof of claim 1.

6. A method for producing an antibody or fragment thereof specifically binding to an extracellularly exposed lysyl-tRNA synthetase (KRS) N-terminal region, the method comprising:

(a) transforming host cells with a recombinant expression vector comprising a polynucleotide encoding the antibody or fragment thereof of claim 1;
(b) incubating the transformed host cells to produce an antibody or fragment thereof; and
(c) collecting the antibody or fragment thereof produced in the host cells.

7. A method for specific detection of an extracellularly exposed lysyl-tRNA synthetase (KRS) N-terminal region, the method comprising:

contacting the antibody or fragment thereof of claim 1 with a sample; and
detecting the antibody or fragment thereof.

8. A method for diagnosing cancer in a subject in need thereof, the method comprising administering the antibody or fragment thereof claim 1 labelled with a detectable moiety to the subject in an amount effective for diagnosing cancer.

Referenced Cited
U.S. Patent Documents
8962560 February 24, 2015 Greene et al.
9511085 December 6, 2016 Kim et al.
20050277157 December 15, 2005 Rose et al.
20130243745 September 19, 2013 Greene
20160377619 December 29, 2016 Kim et al.
20210070880 March 11, 2021 Kwon
Foreign Patent Documents
2012-500256 January 2012 JP
2013532965 August 2013 JP
10-2010-0040583 April 2010 KR
10-2015-0079476 July 2015 KR
2010021415 February 2010 WO
2011153277 December 2011 WO
2019054819 March 2019 WO
Other references
  • Almagro & Fransson, Humanization of antibodies, Frontiers in Bioscience 2008; 13: 1619-33 (Year: 2008).
  • Oct. 1, 20206—(AU) Examination Report—App No. 2018244677.
  • Nov. 4, 2020—(JP) Office Action—App No. 2019-553434.
  • Park et al. “Human lysyl-tRNA synthetase is secreted to trigger proinflammatory response” PNAS, vol. 102, No. 18, May 3, 2005, pp. 6356-6361.
  • Yoshifuji et al. “Anti-aminoacyl-tRNA synthetase antibodies in clinical course prediction of interstitial lung disease complicated with idiopathic inflammatory myopathies” Autoimunity, May 2006; 39(3): 233-241.
  • Aug. 22, 2018 (WO)—International Search Report PCT/KR2018/003594.
  • Kim et al. “Chemical inhibition of prometastic lysyl-tRNA synthetase-maminin receptor interaction” Nature Chemical Biology, vol. 10, Jan. 2014.
  • Jeon et al. “Function of membranous lysyl-tRNA synthetase and its implication for tumorigenesis” Biochimica et Biophysica Acta 1864 (2016) 1707-1713.
  • Dec. 18, 2020 (EP) European Search Report Application No. 18777477.3.
Patent History
Patent number: 11685791
Type: Grant
Filed: Mar 27, 2018
Date of Patent: Jun 27, 2023
Patent Publication Number: 20200102402
Assignee: Zymedi Co., Ltd. (Incheon)
Inventors: Sunghoon Kim (Seoul), Hyunbo Shim (Seoul), Nam Hoon Kwon (Gyeonggi-do), Daeyoung Han (Gyeonggi-do)
Primary Examiner: Peter J Reddig
Assistant Examiner: Cheng Lu
Application Number: 16/498,505
Classifications
Current U.S. Class: Diabetes (514/6.9)
International Classification: C07K 16/40 (20060101); C12N 15/52 (20060101); G01N 33/574 (20060101);